Recombinant Mouse Granulocyte colony-stimulating factor protein (Csf3) (Active)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Mouse Granulocyte Colony-Stimulating Factor (Csf3) Protein (Active)

Recombinant mouse granulocyte colony-stimulating factor (Csf3), also known as G-CSF, is a glycoprotein cytokine engineered to mimic the natural protein's function in hematopoiesis. It regulates granulocyte production, differentiation, and mobilization of hematopoietic stem cells. This recombinant form is produced via bacterial (e.g., E. coli) or mammalian (e.g., HEK 293) expression systems, with variations in post-translational modifications (e.g., glycosylation) affecting functional properties .

Biological Activity and Functional Mechanism

Csf3 binds to its receptor (Csf3r) on hematopoietic progenitors, triggering signaling cascades such as JAK/STAT, MAPK, and PI3K/Akt, which promote neutrophil survival, proliferation, and mobilization .

Dose-Response Data

Assay TypeED50Specific ActivityModel SystemSource
NFS-60 Cell Proliferation≤0.020 ng/mL50 × 10⁶ units/mgDose-dependent proliferation
NFS-60 Cell Proliferation0.01–0.03 ng/mL>1.0 × 10⁷ IU/mgOptimized cell culture

Production and Purification

Recombinant Csf3 is produced via:

  1. Expression:

    • HEK 293: Yields glycosylated, full-length protein (31–208 aa) .

    • E. coli: Produces non-glycosylated, truncated variants (e.g., 179 aa) .

  2. Purification:

    • Chromatography: RP-HPLC, ion-exchange, or affinity chromatography .

    • Quality Control: SDS-PAGE, ESI-TOF mass spectrometry, and endotoxin assays .

Hematopoiesis and Immune Response

  • Neutrophil Mobilization: Csf3 stimulates granulocyte production, critical for infection defense and chemotherapy recovery .

  • Stem Cell Mobilization: Induces hematopoietic stem cell release into the bloodstream for transplantation .

Neurological and Oncological Studies

  • Neuroprotection: Acts as a neurotrophic factor, enhancing neurogenesis and reducing apoptosis in cerebral ischemia models .

  • Cancer Models: Overexpression in urothelial cancers linked to alternative splicing and promoter mutations .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 10 mM Sodium Citrate, pH 4.0, 150 mM NaCl, 0.01 % Tween 20 solution.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our default final concentration of glycerol is 50%, which can be used as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer ingredients, storage temperature, and the stability of the protein itself.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Csf3; CsfgGranulocyte colony-stimulating factor; G-CSF
Datasheet & Coa
Please contact us to get it.
Expression Region
31-208aa
Mol. Weight
18.9 kDa
Protein Length
Full Length of Mature Protein
Purity
>98% as determined by SDS­PAGE.
Research Area
Immunology
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Granulocyte/macrophage colony-stimulating factors are cytokines that play a crucial role in hematopoiesis by regulating the production, differentiation, and function of two related white blood cell populations: granulocytes and monocytes-macrophages. This specific CSF stimulates the production of granulocytes.
Gene References Into Functions
  1. Our research indicates a strong correlation between acute lung injury and elevated serum G-CSF levels, along with increased neutrophil elastase activity in the lungs and serum of mice with adenine-induced acute kidney injury (AKI). G-CSF emerges as a potential target for novel anti-lung injury strategies in patients with AKI. PMID: 30056257
  2. The contrasting roles of G-CSF and IFNgamma in regulating innate inflammatory responses within a murine viral encephalitis model highlight G-CSF as a potential therapeutic target. PMID: 29352287
  3. G-CSF acts as a central regulator in the transition to postinflammatory chronic visceral pain. PMID: 28973941
  4. In aggressive pancreatic ductal adenocarcinoma, elevated G-CSF levels contribute to tumor progression by promoting an increase in the infiltration of neutrophil-like cells exhibiting high immunosuppressive activity. This mechanism suggests a potential avenue for a neoadjuvant therapeutic approach for this challenging disease. PMID: 28775207
  5. Our findings provide compelling evidence for the role of G-CSF in the development of acute chest injury following burn injury. This occurs through the suppression of EPO signaling in bone marrow erythroid cells. PMID: 28867537
  6. Physiologically produced G-CSF not only acts as a neutrophil mobilizer at the relatively late stage of acute inflammation, but also effectively prevents exaggerated neutrophil mobilization. PMID: 27551153
  7. Our data demonstrate that G-CSF is a pivotal driver of disease progression in the K/BxN serum-transfer arthritis (STA) model, likely acting in part by regulating neutrophil numbers in the circulation. PMID: 26848119
  8. SB203580 increases G-CSF expression in macrophages by enhancing the stability of G-CSF mRNA via its 3'UTR. This effect is not attributable to its inhibition of p38 MAPK activity. PMID: 26772539
  9. Our results suggest that G-CSF plays a significant role in preventing colitis, likely through the population of immune regulatory macrophages within the intestine. PMID: 26687628
  10. Our findings provide strong evidence that monophosphoryl lipid A-induced G-CSF facilitates the early expansion, mobilization, and recruitment of neutrophils to the site of infection following burn injury. PMID: 26538529
  11. Overexpression of VEGF may compensate for G-CSF deficiency by preserving cellular components, including blood vessels, in the postinfarction heart. PMID: 25976246
  12. G-CSF rapidly induces autophagy following spinal cord injury to inhibit neuronal apoptosis. PMID: 26524416
  13. G-CSF supports long-term muscle regeneration in mouse models of muscular dystrophy. PMID: 25865621
  14. Constitutive activation of the NF-kappaB pathway in CAIX-depleted cells restored G-CSF secretion. PMID: 25623234
  15. Exposure of Sca-1(+) cells to G-CSF in the culture medium for 72 hours induced time-dependent but self-limiting cell cycle acceleration with a restricted effect on CSC proliferation. PMID: 25160782
  16. Our results suggest that the G-CSF pathway regulates the production of autoantibodies in murine models of lupus. PMID: 23566364
  17. G-CSF acts in a cell-intrinsic manner to expand multipotent progenitors, leading to increased production of tumor-derived Ly6G+ neutrophils. PMID: 25624500
  18. Endothelial cell (EC)-intrinsic MYD88 signaling and subsequent G-CSF production by ECs are essential for myeloid progenitor lineage skewing toward granulocyte-macrophage progenitors, resulting in increased colony-forming unit granulocyte activity. PMID: 24990886
  19. Adventitial CXCL1/granulocyte-colony stimulating factor expression in response to aortic dissection triggers local neutrophil recruitment and activation. This leads to adventitial inflammation via IL-6, ultimately contributing to aortic expansion and rupture. PMID: 25563839
  20. Cultured mouse enteric nervous system -neurospheres exhibit increased expansion with increased G-CSF concentrations, in contrast to central nervous system - derived spheres. PMID: 24253464
  21. The G-CSF STAT3 axis constitutes a key protective mechanism induced by injury to reduce the risk of posttraumatic infection. PMID: 24470495
  22. After deletion of Pten in mice lacking G-CSF, the splenomegaly, myeloproliferative disease, and splenic hematopoietic stem cell accumulation are rescued. PMID: 24127490
  23. Our results suggest that 5-AED survival enhancement is G-CSF-dependent, stimulating innate immune cell function and reducing radiation-induced DNA damage via the induction of genes that modulate cell cycle progression and apoptosis. PMID: 22843381
  24. Activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. PMID: 23530240
  25. G-CSF protein is both necessary and sufficient to restore monocyte chemoattractant protein (MCP)-1 deficiency in neutrophil-mediated host immunity following Klebsiella pneumoniae lung infection. PMID: 23129755
  26. Ang2 deficiency results in enhanced recruitment of myeloid cells, dependent on G-CSF, leading to more aggressive tumor growth and neo-angiogenesis during liver colonization. PMID: 22699974
  27. Il-1rn knockout mice exhibit delayed resolution in acute lung inflammation after exposure to lipopolysaccharide, a process seemingly mediated via the granulocyte colony-stimulating factor/IL-17A axis. PMID: 22592923
  28. Induction of Bv8 expression by granulocyte colony-stimulating factor in CD11b+Gr1+ cells: a key role for Stat3 signaling. PMID: 22528488
  29. Co-administration of BMMCs and G-CSF exhibits synergistic beneficial effects over time. PMID: 21699735
  30. Granulocyte and macrophage populations of murine bone marrow cells are regulated by G-CSF and CD137 protein. PMID: 21179444
  31. TLR2 signaling in G-CSF mobilized PBSCs correlates with their ability to rapidly differentiate into myeloid cells, resulting in improved engraftment. PMID: 21239180
  32. G-CSF is crucial for skeletal myocyte development and regeneration. PMID: 21422169
  33. G-CSF and IL-6 provide signals that determine the angiogenic potential of BM resident monocytes. PMID: 20354107
  34. G-CSF stimulates the expression of the MIP-2 receptor via STAT3-dependent transcriptional activation of Il8rb. PMID: 20185584
  35. G-CSF exerts potent anti-apoptotic activities towards motoneurons in vivo, suggesting that the protection offered by G-CSF in ALS mouse models is due to its direct neuroprotective activity. PMID: 20178614
  36. Overexpression of STAT3beta did not alter the kinetics of G-CSF-mediated neutrophilic differentiation or p27 induction in 32D/G-CSF-R WT cells. PMID: 11920194
  37. Increased production of G-CSF in mice mounting the acute phase response is a key physiological component of host defense. PMID: 12097396
  38. Evaluation of the role of G-CSF in the production, survival, and release of neutrophils from bone marrow into circulation using G-CSF-deficient mice. PMID: 12130495
  39. G-CSF mediates neutrophil mobilization indirectly through the generation of trans-acting signals. PMID: 12387736
  40. G-CSF mediates granulopoiesis and, consequently, participates in neutrophilia in LFA-1 deficient mice. PMID: 12734371
  41. Regulation of G-CSF levels may provide a mechanism for directing primitive hematopoietic progenitors into the common myeloid lineage in response to environmental stresses. PMID: 12893769
  42. G-CSF treatment leads to Lyn-mediated tyrosine phosphorylation of Gab2, which may serve as a crucial intermediate in enhanced Akt activity and myeloid differentiation, rather than growth/survival response. PMID: 14656892
  43. G-CSF signaling in neutrophils is negatively regulated by SOCS3. PMID: 14699146
  44. Observations demonstrate that lipocalin 24p3 is not involved in granulocyte-colony stimulating factor withdrawal-induced apoptosis. PMID: 14703690
  45. A critical role for G-CSF in driving joint inflammation and as a potential therapeutic target in inflammatory joint diseases, such as rheumatoid arthritis. PMID: 15272075
  46. G-CSF induces stabilization of Fli-1 protein in myeloid cells. PMID: 15557108
  47. G-CSF treatment significantly improved survival and liver histology in chemically injured mice, primarily by promoting endogenous repair mechanisms; immunohistochemistry revealed a higher percentage of bone marrow-origin hepatocytes. PMID: 15661404
  48. Febrile-range hyperthermia-induced expression of G-CSF drives the sustained peripheral neutrophilia that occurs during prolonged (36 hours) hyperthermia in a conscious temperature-clamped mouse model. PMID: 15829718
  49. G-CSF modulates angiogenesis by increasing myelomonocytic cells (VEGFR1+ neutrophils) and their release of VEGF. PMID: 16223785
  50. Monocytes mobilized into the bloodstream by G-CSF or AMD3100 stimulate angiogenesis at sites of ischemia through a paracrine mechanism. PMID: 16735597

Show More

Hide All

Database Links

KEGG: mmu:12985

STRING: 10090.ENSMUSP00000037762

UniGene: Mm.1238

Protein Families
IL-6 superfamily
Subcellular Location
Secreted.

Q&A

How does recombinant mouse CSF3 compare functionally to human CSF3?

While both human and mouse CSF3 share functional homology in stimulating granulocyte production, several key differences affect their experimental applications:

Mouse CSF3 acts primarily on mouse cells, though with some cross-reactivity to human cells. Notably, human G-CSF can be used in mouse models such as BDF1 mice, which show a stimulatory response to human G-CSF . This cross-species activity is particularly valuable when evaluating novel delivery methods or fusion proteins.

For experimental design, it's important to note that human G-CSF fusion proteins (like G-CSF-Tf) administered to mice at 5 mg/kg (0.05 μmol/kg) subcutaneously or 50 mg/kg (0.5 μmol/kg) orally have demonstrated effective myelopoietic activity . These dosing parameters can serve as starting points when designing mouse experiments with either human or mouse CSF3 variants.

What are the validated methods for measuring recombinant mouse CSF3 bioactivity?

The standard method for assessing CSF3 bioactivity is the NFS-60 cell proliferation assay. This methodology involves:

  • Washing NFS-60 cells three times with RPMI medium 1640/10% FBS

  • Plating cells in 96-well microtiter plates at 1 × 10^5 cells per ml

  • Adding 10-fold serial dilutions of CSF3 protein

  • Incubating at 37°C in 5% CO2 for 48 hours

  • Performing an MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay:

    • Treating cells with 1 mg/ml MTT in serum-free, phenol red-free medium for 4 hours

    • Dissolving formazan crystals in isopropanol

    • Measuring absorbance at 570 nm

For more advanced applications, researchers can evaluate:

  • STAT3/STAT5 nuclear translocation in transfected cells (e.g., HeLa cells expressing CSF3R)

  • JAK activation via phosphorylation status

  • Receptor internalization and recycling dynamics

  • ROS production in myeloid cells (which involves the Lyn-PI3-kinase-Akt pathway)

How should storage and handling protocols be optimized for maintaining recombinant mouse CSF3 activity?

Recombinant mouse CSF3 requires careful handling as it is "an active protein and may elicit a biological response in vivo" . Optimal storage and handling protocols include:

  • Store at -80°C for long-term storage

  • Prepare single-use aliquots to avoid repeated freeze-thaw cycles

  • Reconstitute in sterile buffer (PBS or manufacturer's recommended buffer)

  • For dilute solutions, add carrier protein (e.g., 0.1% BSA) to prevent adsorption loss

  • For quality control validation, recommended techniques include:

    • SDS-PAGE analysis

    • ESI-TOF mass spectrometry verification (predicted MW: 19008.20 Da)

    • HPLC analysis for purity assessment

What strategies have been developed for enhancing delivery and bioavailability of CSF3?

Recent advances in CSF3 delivery include fusion protein technology that significantly improves administration options:

The G-CSF-transferrin (G-CSF-Tf) fusion protein represents a breakthrough in oral delivery of CSF3. This construct is engineered by fusing human cDNAs encoding G-CSF and transferrin with a dipeptide linker (Leu-Glu) . Unlike chemical conjugation methods that produce heterogeneous mixtures, this recombinant approach yields a consistent product.

Expression in HEK293 cells using protein-free medium allows harvesting of the fusion protein after 5 days, with isolation via ammonium sulfate precipitation. When administered orally, G-CSF-Tf demonstrates sustained myelopoietic effects lasting up to 3 days, compared to the shorter duration of subcutaneously administered native G-CSF .

Administration RouteG-CSF DosageG-CSF-Tf DosageDuration of Effect
Subcutaneous1 mg/kg (0.05 μmol/kg)5 mg/kg (0.05 μmol/kg)Standard duration
Oral10 mg/kg (0.5 μmol/kg)50 mg/kg (0.5 μmol/kg)Up to 3 days

This research demonstrates that transferrin can function as an effective carrier for oral delivery of therapeutic proteins, opening possibilities for other fusion protein constructs with improved bioavailability .

How do mutations in CSF3R affect signaling and what experimental approaches best characterize these alterations?

CSF3R mutations have significant implications for signaling dynamics and are associated with several hematological disorders. Key mutations and their experimental characterization include:

  • Truncation Mutations:

    • Nonsense mutations truncating the carboxyl-terminus of CSF3R occur in approximately 30% of severe congenital neutropenia (SCN) patients

    • These mutations increase to approximately 80% after progression to acute myeloid leukemia (AML)

    • Experimental approach: Compare wild-type and truncated CSF3R signaling in myeloid progenitor proliferation assays

  • Activating Point Mutations:

    • T595I mutation: Found in both SCN/AML and de novo AML patients

    • T617I and T617N mutations: Located in the transmembrane domain

    • These mutations cause ligand-independent activation of CSF3R

  • Experimental Characterization Approaches:

    • Site-directed mutagenesis to create specific mutations (e.g., CSF3R-T595V construct to test hydrophobicity effects)

    • Transfection studies in relevant cell lines

    • Colony formation assays to assess autonomous proliferation

    • Analysis of STAT3/STAT5 nuclear accumulation in the absence of CSF3 stimulation

In mutation studies, substituting threonine for valine at position 595 (T595V) resulted in growth factor-independent progenitor cell proliferation similar to T595I, indicating that hydrophobicity rather than structural changes drives autonomous signaling .

How does recombinant mouse CSF3 contribute to modeling hematological disorders?

Recombinant mouse CSF3 is instrumental in developing models for various hematological conditions:

  • Severe Congenital Neutropenia (SCN) Models:

    • CSF3 is clinically used to treat patients with SCN (neutrophil counts <0.5 million/L)

    • Mouse models with ELANE or HAX1 mutations (common in SCN) can be treated with recombinant CSF3 to study neutrophil response patterns

    • These models help assess how long-term CSF3 therapy might contribute to leukemic progression

  • Leukemia Progression Models:

    • The acquisition of CSF3R mutations represents a significant step in leukemic transformation

    • Studies comparing SCN patients in neutropenia phase versus after progression to AML reveal important insights into mutation accumulation

    • Research shows that CSF3R-T595I mutation was a late event, detected after 17 years of CSF3 therapy in one documented SCN/AML patient

  • Reactive Oxygen Species (ROS) Mechanisms:

    • CSF3-induced ROS production involves the Lyn-PI3-kinase-Akt pathway

    • This mechanism may contribute to genotoxic damage and potential carcinogenesis

    • Mouse models can help elucidate how truncated CSF3R proteins might exacerbate ROS production and DNA damage

What considerations should guide experimental design when studying CSF3 signaling in neutrophil development?

When designing experiments to study CSF3's role in neutrophil development, researchers should consider:

  • Signal Activation and Attenuation Balance:

    • CSF3 signaling requires "a tight but dynamic balance between signal activation and attenuation" to achieve appropriate neutrophil output

    • Experiments should assess both activation pathways and negative feedback mechanisms

    • Time-course studies are essential as signaling dynamics change over time

  • Receptor Expression and Trafficking:

    • Monitor CSF3R expression levels and localization

    • Consider receptor internalization, degradation, and recycling

    • Account for potential expression of truncated or mutant receptors

  • Species-Specific Considerations:

    • While mouse models provide valuable insights, species differences exist

    • For translational research, validation in human cells is recommended

    • BDF1 mice respond to human G-CSF and can serve as useful models

  • Dosing Parameters:

    • Different administration routes require adjusted dosing

    • Subcutaneous: 1 mg/kg (G-CSF) or 5 mg/kg (G-CSF-Tf)

    • Oral: 10 mg/kg (G-CSF) or 50 mg/kg (G-CSF-Tf)

    • Duration of treatment influences outcomes (acute vs. chronic exposure)

  • Readout Selection:

    • Cell proliferation assays (e.g., NFS-60 cells)

    • Colony formation in semi-solid media

    • Flow cytometry for neutrophil maturation stages

    • Signaling pathway activation (JAK-STAT, Lyn-PI3K-Akt)

    • ROS production measurement

What are the common challenges in recombinant mouse CSF3 experiments and how can they be addressed?

Researchers working with recombinant mouse CSF3 frequently encounter several challenges:

  • Activity Loss During Storage:

    • Problem: Decreased bioactivity over time

    • Solution: Store at -80°C in single-use aliquots; add carrier protein for dilute solutions; verify activity via NFS-60 proliferation assay before critical experiments

  • Variable Cellular Response:

    • Problem: Inconsistent proliferation or differentiation results

    • Solution: Characterize CSF3R expression levels in your cell model; ensure cells haven't developed CSF3 independence; verify batch consistency using control cell lines

  • Species Cross-Reactivity Issues:

    • Problem: Unexpected differences when translating between mouse and human systems

    • Solution: Validate cross-species activity before experimental design; consider species-specific receptors and downstream signaling differences

  • Mutation Detection Challenges:

    • Problem: Difficulty identifying CSF3R mutations

    • Solution: Use denaturing high-performance liquid chromatography (dHPLC) followed by Sanger sequencing of samples showing aberrant patterns

How can researchers verify the structural integrity and activity of recombinant mouse CSF3 preparations?

Quality control for recombinant mouse CSF3 requires multiple complementary approaches:

  • Structural Verification:

    • Mass spectrometry (ESI-TOF) to confirm molecular weight (expected: 19008.20 ±10 Da)

    • SDS-PAGE analysis to assess purity (should be ≥95%)

    • Western blotting using specific antibodies against mouse CSF3

  • Functional Validation:

    • NFS-60 cell proliferation assay (gold standard for bioactivity)

    • MTT assay following 48-hour incubation with serial dilutions

    • Comparison to a reference standard with known activity

  • Purity Assessment:

    • HPLC analysis to confirm absence of contaminants

    • Endotoxin testing (should be ≤0.005 EU/μg)

    • Sterility testing for cell culture applications

  • Post-Translational Modification Analysis:

    • Verification of O-glycosylation patterns

    • Assessment of proper disulfide bond formation

    • Confirmation of correct protein folding via circular dichroism or other structural techniques

By implementing these comprehensive quality control measures, researchers can ensure consistent and reliable results when working with recombinant mouse CSF3 protein in their experimental systems.

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.