Recombinant Mouse Granulocyte-macrophage colony-stimulating factor (Csf2) (Active)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mouse Granulocyte-macrophage Colony-Stimulating Factor (Csf2) (Active)

Recombinant Mouse Granulocyte-macrophage Colony-Stimulating Factor (Csf2) (Active) is a bioengineered cytokine produced via bacterial expression systems, primarily Escherichia coli. It functions as a growth factor for hematopoietic cells, promoting the differentiation and proliferation of granulocytes and macrophages . This recombinant protein is structurally and functionally analogous to the naturally occurring GM-CSF, which is critical for immune system development and defense against infections .

Amino Acid Sequence and Molecular Features

The recombinant mouse GM-CSF spans amino acids 18–141 (Ala18-Lys141), with a monomeric structure and a molecular weight of ~14.3 kDa . Key structural details include:

ParameterValueSource
Amino Acid SequenceMAPTRSPITVTRPWKHVEAIKEALNLLDDMPVTLNEEVEVVSNEFSFKKLTCVQTRLKIF EQGLRGNFTKLKGALNMTASYYQTYCPPTPETDCETQVTTYADFIDSLKTFLTDIPFECKKPVQK
Molecular Weight14.3 kDa
Glycosylation StatusNonglycosylated (bacterial origin)
Purity>95% (SDS-PAGE)
Endotoxin Level<1.0 EU/μg

Functional Roles

Recombinant mouse GM-CSF activates hematopoietic progenitors via the GM-CSF receptor (CD116/CD131), triggering signaling cascades involving STAT5 and STAT3 . Key activities include:

  • Stimulation of granulocyte/macrophage colonies: Supports differentiation of bone marrow progenitors into neutrophils, eosinophils, and monocytes .

  • Enhancement of immune cell functions: Increases neutrophil migration, phagocytosis, and reactive oxygen species production .

  • Modulation of inflammatory responses: Induces pro-inflammatory cytokines (e.g., TNFα, CCL17) and microglial activation in the CNS .

Bioactivity Assays

Assay TypeResultsSource
FDC-P1 Cell ProliferationED50: 5–50 pg/mL (indicative of granulocyte/macrophage colony formation)
DA3 Cell ProliferationED50: 5–30 pg/mL (myeloma cell growth)
Microglial ActivationIncreases CD40, TNFα, and CD11c mRNA expression in murine brain models

Immunology and Neuroscience

  • Neuroinflammation studies: Anti-GM-CSF antibodies suppress microglial activity in Alzheimer’s disease and multiple sclerosis models .

  • Cancer immunotherapy: Enhances tumor antigen presentation by dendritic cells and modulates Th1/Treg balance .

Infectious Disease Models

  • Pulmonary aspergillosis: Intranasal GM-CSF reduces Aspergillus burden in immunocompromised mice .

  • Bacterial/viral infections: Augments neutrophil recruitment and fungal zinc deprivation mechanisms .

Potential Therapeutic Targets

ApplicationMechanismSource
Neutropenia managementRestores neutrophil production post-chemotherapy (e.g., sargramostim)
Rheumatoid arthritisAnti-GM-CSF antibodies (e.g., otilimab) reduce synovial inflammation
COVID-19 and colitisMonoclonal antibodies under trial to mitigate hyperinflammatory states

Challenges and Considerations

  • Species specificity: Mouse GM-CSF lacks activity on human cells, necessitating species-matched models .

  • Endotoxin control: Critical for in vivo studies to avoid confounding inflammatory responses .

Comparative Analysis of Recombinant GM-CSF Preparations

ParameterMouse GM-CSF (E. coli)Human GM-CSF (CHO Cells)
SourceE. coliMammalian cells
GlycosylationNonglycosylatedGlycosylated
Receptor BindingMouse-specificHuman-specific
Purity>95%>95%
Typical UseMurine modelsHuman cell culture

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 1xPBS, pH 7.4.
Form
Lyophilized powder
Lead Time
Generally, we can ship the products within 5-10 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery time, please consult your local distributors.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging this vial prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use this as a reference.
Shelf Life
The shelf life is influenced by multiple factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. For lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal 6xHis-tagged
Synonyms
Csf2; Csfgm; Granulocyte-macrophage colony-stimulating factor; GM-CSF; Colony-stimulating factor; CSF
Datasheet & Coa
Please contact us to get it.
Expression Region
18-141aa
Mol. Weight
15.1 kDa
Protein Length
Full Length of Mature Protein
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Immunology
Source
Mammalian cell
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Cytokine that stimulates the growth and differentiation of hematopoietic precursor cells from various lineages, including granulocytes, macrophages, eosinophils and erythrocytes.
Gene References Into Functions
  1. Data show that the microbiota enhances respiratory defenses via granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling, which stimulates pathogen killing and clearance by alveolar macrophages. PMID: 29142211
  2. The data indicate that GM-CSF drives chronic tissue damage and disability in experimental autoimmune encephalomyelitis via pleiotropic pathways, but it is dispensable during early lesion formation and the onset of neurologic deficits. PMID: 29288202
  3. The release of IL-33 and GM-CSF from epithelial cells induces the activation of p65 and the p38-MK2/3 signaling module in Dendritic Cells, resulting in Th2 polarization and, ultimately, allergic inflammation. PMID: 29288203
  4. Results show T cell production of GM-CSF contributes to control of M. tuberculosis infection in the absence of other sources of GM-CSF, that multiple T cell subsets make GM-CSF in the lung over the course of infection and that GM-CSF can act directly on infected macrophages through a pathway requiring PPARgamma to limit bacterial growth. PMID: 29066547
  5. In conclusion, our study confirms the pathogenic role of GM-CSF in colitis-associated colorectal cancer development. GM-CSF favors tumor-permissive microenvironment by inducing MDSC generation and recruiting them into colonic tissues. PMID: 28534709
  6. These data demonstrate that GM-CSF levels during radiotherapy can be used as a prognostic biomarker for lung and esophageal cancer. PMID: 27835886
  7. This study demonstrates that epithelial-derived GM-CSF is a critical early signal during allergic sensitization to cockroach allergen. PMID: 27731325
  8. These impaired macrophage functions in leukemic mice were significantly corrected by IL-3 and GM-CSF treatment, indicating the therapeutic benefit of these two cytokines in leukemia. PMID: 28039843
  9. Both IL-6 protein production and transcript levels were downregulated by RA in respiratory tract epithelial cells (LETs), but upregulated in macrophages (MACs). RA also increased transcript levels of MCP-1, GMCSF, and IL-10 in MACs, but not in LETs. Conversely, when LETs, but not MACs, were exposed to RA. PMID: 27940088
  10. T-GM-CSF and -IL-3 significantly, and reciprocally, blunted receptor binding and myeloid progenitor cell proliferation activity of both FL-GM-CSF and -IL-3 in vitro and in vivo. PMID: 28344320
  11. Results indicate GM-CSF as both a key contributor to the pathogenesis of MI and a potential therapeutic target. PMID: 28978634
  12. Obesity alters the lung neutrophil infiltration to enhance breast cancer metastasis through IL5 and GM-CSF. PMID: 28737771
  13. GM-CSF is required for the normal balance of leukocyte subsets, including granulocytes, B cells, and naive vs. effector T cells. There was an approximately 3-fold increase in the percentages of granulocytes in Csf2-/- PBMCs. The presence of maximal experimental autoimmune encephalomyelitis in the complete absence of GM-CSF revealed that GM-CSF is not an obligate effector molecule in all forms of EAE. PMID: 27256565
  14. chemerin inhibited nuclear factor-kappaB activation and the expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-6) by tumor cells and tumor-associated endothelial cell, respectively, via its receptors, and consequently, MDSC induction was impaired, leading to restoration of antitumor T-cell response and decreased tumor angiogenesis. PMID: 28166197
  15. These findings describe a novel role for GM-CSF as an essential initiating cytokine in cardiac inflammation. PMID: 27595596
  16. Data reviewed establish that any damage to brain tissue tends to cause an increase in G-CSF and/or GM-CSF (G(M)-CSF) synthesized by the brain. Glioblastoma cells themselves also synthesize G(M)-CSF. G(M)-CSF synthesized by brain due to damage by a growing tumor and by the tumor itself stimulates bone marrow to shift hematopoiesis toward granulocytic lineages away from lymphocytic lineages. PMID: 28459367
  17. Evi1(+)DA-3 cells modified to express an intracellular form of GM-CSF, acquired growth factor independence and transplantability and caused an overt leukemia in syngeneic hosts, without increasing serum GM-CSF levels. PMID: 25907616
  18. IL-23-induced GM-CSF mediates the pathogenicity of CD4(+) T cells in experimental autoimmune myocarditis. PMID: 26660726
  19. GM-CSF accelerated the G1/S phase transition in EPCs by upregulating the expression of cyclins D1 and E. PMID: 24662605
  20. host RNF13 affects the concentration of GM-CSF in tumor-bearing lungs. PMID: 26197965
  21. Sc CW-derived BG stimulated the late and strong expression of Csf2 in a dectin-1-dependent manner, they remain poor inducers of chemokine and cytokine production in murine macrophages. PMID: 26840954
  22. GM-CSF and uPA are required for Porphyromonas gingivalis-induced alveolar bone loss in a mouse periodontitis model. PMID: 25753270
  23. These findings identify GM-CSF as central to the protective immune response that prevents progressive fungal disease. PMID: 26755822
  24. regulatory molecule causative of aortic dissection and intramural haematoma. PMID: 25923510
  25. Results indicate that granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling controls a pathogenic expression signature in CC chemokine receptors 2 (CCR2+)Ly6C(hi) monocytes. PMID: 26341401
  26. TL1A increases expression of CD25, LFA-1, CD134 and CD154, and induces IL-22 and GM-CSF production from effector CD4 T-cells. PMID: 25148371
  27. Mafb-deficient microglia cultured with GM-CSF barely extended their membrane protrusions, probably due to abnormal activation of RhoA, a key regulator of cytoskeletal remodeling. MafB is a negative regulator of GM-CSF signaling in microglia. PMID: 25998393
  28. Csf2-/- mice showed a 30% increase in whole body adiposity, which persisted to adulthood. Adipocytes from Csf2-/- mice were 50% larger by volume and contained higher levels of adipogenesis gene transcripts, indicating enhanced adipocyte differentiation. PMID: 25931125
  29. Renal ischemia reperfusion injury tubular cells expressed elevated GM-0CSF, that supports tubular cells proliferation. PMID: 25388222
  30. GM-CSF is key to the development of experimental osteoarthritis and its associated pain. PMID: 22995428
  31. Loss of encephalitogenic activity of STAT5-deficient autoreactive CD4+ T cells was independent of IFN-gamma or IL-17 production, but was due to the impaired expression of GM-CSF, a crucial mediator of T-cell pathogenicity. PMID: 25412660
  32. These data indicate that GM-CSF plays a role in the inflammatory signaling network that drives neutrophil recruitment in response to Clostridium difficile infection but does not appear to play a role in clearance of the infection. PMID: 25045999
  33. IRF8 represses GM-CSF expression in T cells to affect myeloid cell lineage differentiation. PMID: 25646302
  34. Reprogramming of monocytes by GM-CSF contributes to regulatory immune functions during intestinal inflammation. PMID: 25653427
  35. GM-CSF promotes advanced plaque progression by increasing macrophage apoptosis susceptibility. PMID: 25348165
  36. although iTreg cells maintained the ability to produce IFN-gamma and TNF-alpha in vivo, their ability to produce GM-CSF was selectively degraded upon antigen stimulation under inflammatory conditions. PMID: 25168419
  37. IFN-gamma has a protective role in the demyelination of brain through downregulation of IL-17/GM-CSF and induction of neuroprotective factors in the brain by monocytes/microglial cells. PMID: 25339670
  38. Expression of PPAR-gamma in fetal lung monocytes was dependent on the cytokine GM-CSF. Therefore, GM-CSF has a lung-specific role in the perinatal development of alveolar macrophages through the induction of PPAR-gamma in fetal monocytes. PMID: 25263125
  39. These novel findings indicate that the inflammatory mediator, GM-CSF, may have significant protective properties in the chronic sequelae of experimental Traumatic brain injury. PMID: 24392832
  40. Airway infections and pollutants increase the susceptibility to develop allergic asthma through a GM-CSF/IL-33/OX40L pathway. PMID: 24551140
  41. Data indicate that neutralization of GM-CSF would be a useful therapeutic strategy for severe connective tissue diseases (CTD)-interstitial lung disease (ILD). PMID: 24951817
  42. Data indicate that lung epithelium-derived GM-CSF is a critical regulator of CD11b+ dendritic cells (DCs)-mediated Th2 cell priming. PMID: 24943219
  43. Bhlhe40 is required positively regulates the production of GM-CSF and negatively regulates the production of IL-10 in T cells. PMID: 24699451
  44. Report IL-18 plus GM-CSF modified tumor cell vaccine can induce significant antitumor effects in Lewis lung carcinoma. PMID: 24475975
  45. Data indicate that in type 1 diabetes monocytes STAT5Ptyr activation is significantly higher and is found bound to CSF2 promoter and PTGS2 enhancer regions. PMID: 24204704
  46. Data indicate that GM-CSF controls IgM production in innate response activator B cells. PMID: 24821911
  47. our findings suggest that over-production of GM-CSF by T cells could be a pathogenic factor in many histiocytic disorders. PMID: 24183847
  48. that p52 binds to the promoter of the GM-CSF-encoding gene (Csf2) and cooperates with c-Rel in the transactivation of this target gene. PMID: 24899500
  49. GM-SCF, IL-21 and Rae1 expression, alone or in combination, induces a cellular immune response against H22 tumor cells. PMID: 24350772
  50. Data indicate that GM-CSF knockout mice exhibit a unique mix of M1-M2 macrophage phenotypes in alveolar macrophages. PMID: 24044676

Show More

Hide All

Database Links

KEGG: mmu:12981

STRING: 10090.ENSMUSP00000019060

UniGene: Mm.4922

Protein Families
GM-CSF family
Subcellular Location
Secreted.

Q&A

What is Recombinant Mouse GM-CSF and what are its fundamental properties?

Recombinant Mouse Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF, also designated as Csf2) is a hematopoietic growth factor initially characterized for its ability to support in vitro colony formation of granulocyte-macrophage progenitors . It is a 14.3 kDa glycoprotein consisting of 125 amino acids that functions as a monomer . The amino acid sequence of mouse GM-CSF is: MAPTRSPITVTRPWKHVEAIKEALNLLDDMPVTLNEEVEVVSNEFSFKKLTCVQTRLKIFEQGLRGNFTKLKGALNMTASYYQTYCPPTPETDCETQVTTYADFIDSLKTFLTDIPFECKKPVQK .

Recombinant mouse GM-CSF is typically produced in E. coli expression systems to ensure proper folding and biological activity, with high purity (≥95%) as determined by reducing and non-reducing SDS-PAGE . Its biological activity is commonly measured using proliferation assays with FDC-P1 or FDCP-1 cell lines, with typical ED₅₀ values less than 50 pg/ml, corresponding to a specific activity of approximately 2 × 10⁷ units/mg .

How does mouse GM-CSF compare to human GM-CSF in experimental systems?

Mouse and human GM-CSF exhibit significant species specificity with no cross-reactivity, making it essential to use the appropriate species-matched reagent in experimental systems . This species specificity stems from substantial differences in their amino acid sequences and tertiary structures, despite sharing similar biological functions within their respective species.

When designing experiments involving xenografts or humanized mouse models, researchers must account for this species specificity. For instance, in human cell lines or primary cells transplanted into mouse models, supplementation with human GM-CSF rather than mouse GM-CSF is necessary to support the human cells. Conversely, when studying mouse myeloid cells, only mouse GM-CSF will effectively stimulate the appropriate signaling pathways .

The receptor for GM-CSF consists of a ligand-binding α-chain and a signal-transducing β-chain (CSF2Rβ or CSF2 receptor subunit beta), with both components being required for proper signaling . This receptor complex shows similar organization between species but differs in sequence, explaining the lack of cross-reactivity.

What cell types produce and respond to GM-CSF in mouse models?

In mouse models, GM-CSF is produced by numerous cell types in response to inflammatory stimuli or cytokine signaling. These producer cells include:

  • T lymphocytes (particularly activated T cells)

  • B lymphocytes

  • Macrophages

  • Mast cells

  • Endothelial cells

  • Fibroblasts

  • Adipocytes

The cellular targets of GM-CSF include both progenitor and mature cells of the myeloid lineage:

  • Progenitor cells: GM-CSF supports the development and proliferation of:

    • Granulocyte-macrophage progenitors

    • Erythroid progenitors

    • Megakaryocyte progenitors

    • Eosinophil progenitors

  • Mature cells: GM-CSF acts as a survival factor and activates the effector functions of:

    • Granulocytes

    • Monocytes/macrophages

    • Eosinophils

In inflammatory settings, increased local production of GM-CSF dramatically alters myeloid cell function, promoting inflammatory cytokine production, antigen presentation capabilities, and migration to sites of inflammation . This is particularly important in models of autoimmune disease, where GM-CSF significantly contributes to pathogenesis by modulating myeloid cell behavior rather than directly affecting myelopoiesis .

What is the optimal methodology for storing and reconstituting recombinant mouse GM-CSF?

For optimal maintenance of biological activity, recombinant mouse GM-CSF requires careful handling and storage conditions:

Storage recommendations:

  • Lyophilized protein should be stored at -20°C or -80°C

  • Avoid repeated freeze-thaw cycles

  • Protect from light and moisture

  • Monitor expiration dates carefully

Reconstitution protocol:

  • Allow the lyophilized protein to reach room temperature

  • Reconstitute in sterile water to a concentration of 0.1-1.0 mg/ml

  • Gently swirl or rotate the vial until complete dissolution (avoid vigorous shaking which can cause protein denaturation)

  • Once reconstituted, prepare single-use aliquots

  • Store reconstituted aliquots at -20°C or -80°C

  • For working solutions, dilute in appropriate cell culture medium containing carrier protein (0.1-1% BSA or serum)

Stability considerations:

  • Reconstituted GM-CSF maintains activity for approximately 1-2 weeks at 4°C

  • For longer storage, aliquot and freeze at -20°C or -80°C

  • Avoid more than 3 freeze-thaw cycles

  • Working solutions should be prepared fresh for each experiment

The endotoxin level should be ≤1.00 EU/μg to prevent non-specific activation of cells in experimental systems, particularly when studying inflammatory responses .

How can recombinant mouse GM-CSF be utilized in mouse models of inflammatory diseases?

Recombinant mouse GM-CSF serves as both an investigative tool and a therapeutic target in inflammatory disease models. Its application extends to several experimental approaches:

As an investigative tool:

  • In vivo administration: Direct injection of recombinant GM-CSF can be used to study its effects on inflammatory disease progression. Typically administered at 0.1-10 μg per mouse, depending on the model and desired effect .

  • Ex vivo cell priming: Monocytes or macrophages can be primed with GM-CSF (typically 10 ng/ml) before adoptive transfer or further stimulation. This approach has revealed that GM-CSF priming significantly enhances the IL-1β production by anti-MPO stimulated monocytes, demonstrating how GM-CSF modifies myeloid cell responses in autoimmune contexts .

  • Cell differentiation: GM-CSF is used to generate bone marrow-derived dendritic cells (BMDCs) or specific macrophage phenotypes in vitro, which can then be used for mechanistic studies or adoptive transfer experiments.

As a therapeutic target:

  • Neutralizing antibodies: Anti-GM-CSF antibodies have shown efficacy in reducing kidney damage in the nephrotoxic nephritis (NTN) model, highlighting GM-CSF's pathogenic role in crescentic glomerulonephritis .

  • Receptor targeting: Studies using CSF2rb-deficient mice have demonstrated protection from anti-MPO induced glomerulonephritis, even without reduction in myeloid cell infiltration, suggesting that GM-CSF signaling modifies cellular function rather than recruitment .

  • Genetic approaches: GM-CSF knockout mice or conditional deletion models provide insights into disease pathogenesis, though these approaches must account for potential developmental effects.

In antineutrophil cytoplasmic antibody-associated vasculitis (AAV) models, GM-CSF expression increases significantly in kidney tissue and urine, correlating positively with the percentage of renal crescents . This suggests that monitoring GM-CSF levels could serve as a biomarker for disease activity.

What mechanisms underlie GM-CSF's role in modulating T cell responses in inflammatory conditions?

GM-CSF significantly influences T cell responses through both direct and indirect mechanisms, particularly in inflammatory and autoimmune settings:

Indirect mechanisms via myeloid cells:

  • Enhanced antigen presentation: GM-CSF-stimulated monocytes and dendritic cells upregulate MHC class II and co-stimulatory molecules, enhancing their ability to present antigens to T cells.

  • Cytokine production modulation: When primed with GM-CSF (10 ng/ml), monocytes stimulated with anti-MPO antibodies produce significantly more IL-1β than unprimed cells . This altered cytokine environment directly influences T cell polarization.

  • Induction of Th17 polarization: Experimental data demonstrate that supernatants from GM-CSF-primed, anti-MPO-stimulated monocytes strongly induce Th17 polarization in vitro . This mechanism appears critical in vivo, as CSF2rb-deficient chimeric mice show reduced numbers of kidney-infiltrating Th17 cells in AAV models .

Experimental approach to study this phenomenon:

  • Isolate bone marrow-derived monocytes from mice

  • Prime one group with recombinant mouse GM-CSF (10 ng/ml) for 6-24 hours

  • Stimulate both primed and unprimed monocytes with anti-MPO IgG

  • Collect supernatants after 24 hours

  • Add supernatants to cultures of CD4+ T cells isolated from mouse spleens

  • After 5 days, analyze T cell polarization by flow cytometry (particularly focusing on IL-17A production)

  • Compare Th17 polarization between conditions

This experimental paradigm has revealed that GM-CSF fundamentally alters the capacity of monocytes to influence adaptive immunity, creating a feed-forward inflammatory loop in autoimmune conditions like AAV. Understanding these mechanisms provides potential intervention points for therapeutic development.

How do experimental models utilizing GM-CSF receptor deficiency differ from GM-CSF neutralization approaches?

The experimental approaches targeting GM-CSF function can be categorized into receptor-based and ligand-based strategies, each with distinct advantages and limitations:

GM-CSF receptor deficiency models:

  • Complete CSF2rb knockout: Mice lacking the CSF2rb subunit (β chain) of the GM-CSF receptor demonstrate protection from anti-MPO-induced glomerulonephritis, with significantly reduced crescent formation compared to wild-type mice . These models reveal the importance of GM-CSF signaling in disease pathogenesis while maintaining normal baseline myelopoiesis.

  • Cell-specific receptor deletion: More sophisticated models using Cre-loxP systems allow deletion of CSF2rb in specific cell populations. For example, deletion in CCR2+ monocytes protected mice from experimental autoimmune encephalomyelitis, demonstrating cell-specific roles for GM-CSF signaling .

  • Chimeric approaches: Irradiating MPO-deficient mice and reconstituting with CSF2rb-deficient bone marrow creates chimeric models that allow specific investigation of the role of GM-CSF signaling in hematopoietic cells while maintaining normal receptor expression in non-hematopoietic tissues .

GM-CSF neutralization approaches:

  • Anti-CSF2 antibodies: Direct neutralization of GM-CSF protein using monoclonal antibodies provides temporal control over GM-CSF inhibition. This approach reduced kidney damage in nephrotoxic nephritis models, with decreased crescentic and necrotic glomeruli compared to isotype controls .

  • Soluble receptor administration: Recombinant soluble GM-CSF receptor can act as a decoy, binding and sequestering GM-CSF.

  • Small molecule inhibitors: Various compounds targeting the GM-CSF receptor or downstream signaling pathways.

Comparative analysis:

ParameterReceptor DeficiencyNeutralizing Antibodies
Temporal controlLimited (usually constitutive)High (can administer at different disease stages)
SpecificityHigh tissue/cell specificity with conditional modelsDepends on antibody distribution and half-life
Developmental effectsPotential compensatory mechanismsMinimal when used in adult animals
Degree of inhibitionComplete in knockout cellsDose-dependent, potentially incomplete
Experimental complexityHigher (requires genetic manipulation)Lower (simple administration protocols)
Translational relevanceLess direct application to human therapeuticsMore closely mimics clinical therapeutic approaches

Both approaches have revealed that GM-CSF signaling is critical for developing crescentic glomerulonephritis in mouse models, with the mechanism involving altered monocyte function and subsequent T cell polarization rather than changes in myeloid cell recruitment .

What methodological considerations are important when using GM-CSF for in vitro differentiation of myeloid cells?

When utilizing GM-CSF for in vitro differentiation of mouse myeloid cells, several methodological considerations are crucial for experimental success and reproducibility:

Starting material optimization:

  • Bone marrow isolation technique: The femur and tibia should be flushed with cold PBS under sterile conditions. For enhanced yield, consider including additional bones such as the pelvis.

  • Cell quality assessment: Viability should exceed 90% before initiating cultures; lower viability may compromise differentiation efficiency.

  • Red blood cell lysis: Gentle lysis using ammonium chloride-based buffers is preferred to prevent damage to progenitor cells.

Culture conditions:

  • Recombinant GM-CSF concentration:

    • For dendritic cell differentiation: 20-40 ng/ml

    • For macrophage differentiation: 10-20 ng/ml

    • Higher concentrations may not improve yield and can alter phenotypes

  • Culture duration:

    • Dendritic cells: 6-8 days

    • Macrophages: 5-7 days

    • Extended culture periods may result in spontaneous activation

  • Media selection:

    • RPMI-1640 or DMEM supplemented with 10% FBS

    • Consider using defined serum replacements for higher reproducibility

    • Antibiotics (penicillin/streptomycin) should be included to prevent contamination

  • Feeding schedule:

    • Add fresh GM-CSF-containing medium every 2-3 days

    • For dendritic cells, consider partial medium replacement to retain non-adherent and loosely adherent cells

Validation of differentiated cells:

  • Flow cytometry markers:

    • GM-CSF-derived dendritic cells: CD11c+, MHC-II+, CD86+

    • GM-CSF-derived macrophages: F4/80+, CD11b+

  • Functional assays:

    • Phagocytosis capacity

    • Cytokine production in response to TLR ligands

    • T cell stimulation capacity

Important technical considerations:

  • Loss of MPO expression: Human monocyte-derived macrophages lose MPO protein expression during differentiation with either GM-CSF or M-CSF, rendering them unresponsive to anti-MPO ANCA activation . This transformation should be considered when designing ANCA-related experiments.

  • Batch testing: Each new lot of recombinant GM-CSF should be tested for biological activity using standardized assays, such as FDC-P1 cell proliferation, before use in critical experiments .

  • Endotoxin contamination: Ensure the recombinant protein has low endotoxin levels (≤1.00 EU/μg) to prevent non-specific activation, especially when studying inflammatory responses .

  • Species matching: Remember that mouse and human GM-CSF show no cross-reactivity; always use species-appropriate cytokines .

What are common issues in GM-CSF-based experiments and their solutions?

Researchers working with recombinant mouse GM-CSF frequently encounter challenges that can impact experimental outcomes. The following troubleshooting guide addresses common issues and provides evidence-based solutions:

Problem: Low biological activity in functional assays

Potential causes and solutions:

  • Protein denaturation: GM-CSF is sensitive to improper handling. Avoid vigorous shaking, repeated freeze-thaw cycles, and extended storage at 4°C. Use siliconized tubes for dilution and storage .

  • Carrier protein absence: GM-CSF may adhere to plastic surfaces. Include 0.1-1% BSA or serum in working solutions to prevent adsorption .

  • Incorrect reconstitution pH: Ensure reconstitution buffer is at physiological pH (7.2-7.4). Acidic or basic conditions can reduce activity.

  • Endotoxin contamination: High endotoxin levels can interfere with many assays. Verify that endotoxin levels are ≤1.00 EU/μg as measured by LAL assay .

Problem: Variable cell yields in differentiation protocols

Potential causes and solutions:

  • Starting cell population heterogeneity: Consider enriching for progenitor cells using negative selection before initiating cultures.

  • Inconsistent GM-CSF dosing: Prepare master stocks at high concentration and make single-use aliquots to ensure consistent dosing across experiments.

  • Serum batch variation: Serum components can significantly affect differentiation. Pre-screen serum batches or switch to serum-free formulations with defined supplements.

  • Cell density effects: Maintain consistent seeding density (typically 1-2×10⁶ cells/ml) as both over-crowding and under-seeding can affect differentiation.

Problem: Unexpected inflammatory activation

Potential causes and solutions:

  • Endotoxin contamination: Even low levels of endotoxin can activate cells, particularly when studying inflammatory mechanisms. Use endotoxin-tested reagents throughout .

  • Cell stress during isolation: Minimize processing time and maintain cells at appropriate temperatures to reduce stress-induced activation.

  • Mycoplasma contamination: Routinely test cultures for mycoplasma, which can cause aberrant activation patterns.

Problem: Failure to observe expected GM-CSF effects in vivo

Potential causes and solutions:

  • Dosing inadequacy: Mouse experiments typically require 0.1-10 μg per mouse, depending on the model. Consider dose-response studies to determine optimal concentration .

  • Timing of administration: GM-CSF effects are context-dependent. Administration relative to disease induction or inflammatory stimulus is critical.

  • Strain differences: Background strain can significantly impact GM-CSF responsiveness. C57BL/6 and BALB/c mice may show different responses to the same dose.

  • Neutralizing antibodies: Some mouse strains may develop antibodies against recombinant proteins, particularly with repeated dosing. Consider testing for anti-GM-CSF antibodies in serum.

How can researchers effectively measure GM-CSF levels in experimental samples?

Accurate quantification of GM-CSF in experimental samples is essential for understanding its role in various physiological and pathological processes. Multiple methodologies are available, each with specific advantages for different sample types and research questions:

Enzyme-Linked Immunosorbent Assay (ELISA)

Methodology:

  • Commercial kits typically use a sandwich ELISA format with detection limits of 1-5 pg/ml

  • Suitable for serum, plasma, cell culture supernatants, and tissue lysates

  • Sample preparation varies by source:

    • Serum/plasma: Centrifugation to remove cellular components

    • Tissue: Homogenization in appropriate buffer with protease inhibitors

    • Urine: Centrifugation and potential concentration for dilute samples

Research evidence:
GM-CSF protein detection in kidney lysates using ELISA revealed high levels in AAV mice (approximately 75-100 pg/mg tissue) but undetectable levels in control mice . Similarly, urinary GM-CSF was significantly elevated in AAV mice but undetectable in healthy controls .

Flow Cytometry-Based Bead Arrays

Methodology:

  • Multiplex bead-based assays allow simultaneous detection of multiple cytokines including GM-CSF

  • Advantages include smaller sample volumes and multiplexing capability

  • Typically sensitive to 5-10 pg/ml

  • Particularly useful for serum and cell culture supernatants with limited volume

In situ Hybridization for GM-CSF mRNA

Methodology:

  • RNAscope or similar technologies can detect GM-CSF mRNA in tissue sections

  • Provides spatial information about GM-CSF-producing cells

  • Sample preparation involves tissue fixation, sectioning, and probe hybridization

Research evidence:
In situ hybridization demonstrated upregulation of CSF2 expression in glomerulus-infiltrating cells in kidney biopsies from patients with active AAV . This technique revealed the cellular source of GM-CSF within the tissue microenvironment.

Quantitative PCR for Csf2 gene expression

Methodology:

  • Extract RNA from tissues or cells using standard protocols

  • Generate cDNA through reverse transcription

  • Perform qPCR with validated primers specific for mouse Csf2

  • Normalize to appropriate housekeeping genes (e.g., GAPDH, β-actin)

Research evidence:
Kidney Csf2 mRNA expression was strongly upregulated in AAV mice compared to controls, confirming local expression at the transcriptional level .

Considerations for sample collection and processing:

  • Timing: GM-CSF has a relatively short half-life in circulation. Standardize collection times relative to treatment or disease stage.

  • Protease inhibitors: Include protease inhibitors in all sample preparation buffers to prevent degradation.

  • Sample storage: Process samples immediately or store at -80°C. Avoid repeated freeze-thaw cycles.

  • Assay validation: Always include appropriate positive and negative controls. Consider spike-recovery experiments for complex biological matrices.

  • Standard curves: Use recombinant mouse GM-CSF to generate standard curves in the same matrix as experimental samples when possible.

What are the current experimental approaches to study GM-CSF signaling pathways in mouse models?

Investigating GM-CSF signaling pathways requires integrated approaches spanning from molecular to systems levels. Contemporary experimental strategies include:

Genetic Modification Approaches

  • Receptor subunit knockouts: Studies using CSF2rb-deficient mice have demonstrated protection from anti-MPO induced glomerulonephritis, revealing the importance of GM-CSF receptor signaling in disease pathogenesis .

  • Conditional deletion systems: Cell-specific deletion of CSF2rb using Cre-loxP technology enables investigation of GM-CSF signaling in specific cell populations. For example, deletion in CCR2+ monocytes protected mice from experimental autoimmune encephalomyelitis .

  • CRISPR/Cas9 gene editing: This approach allows precise modification of signaling components with reduced off-target effects compared to traditional knockout methods.

Phosphoproteomic Analysis

  • Western blotting for signaling proteins: Monitor phosphorylation of key downstream mediators including:

    • JAK2/STAT5 pathway

    • MAP kinase pathways (ERK1/2)

    • PI3K/Akt pathway

    • NF-κB pathway

  • Mass spectrometry-based approaches: Provides comprehensive analysis of phosphorylation events following GM-CSF stimulation, revealing novel signaling components and pathway interactions.

  • Phospho-flow cytometry: Enables single-cell analysis of phosphorylation events in heterogeneous cell populations, particularly useful for ex vivo analysis of signaling in primary cells.

Pharmacological Inhibition Studies

  • JAK inhibitors: Compounds like ruxolitinib inhibit JAK2, a critical mediator of GM-CSF signaling.

  • PI3K inhibitors: Wortmannin or LY294002 block PI3K-dependent events downstream of GM-CSF receptor activation.

  • MEK/ERK inhibitors: PD98059 or U0126 inhibit MAPK pathway activation.

  • Small molecule GM-CSF receptor antagonists: Directly interfere with ligand-receptor binding or receptor dimerization.

Transcriptomic Analysis

  • RNA-sequencing: Provides comprehensive analysis of gene expression changes induced by GM-CSF in different cell types.

  • ChIP-seq for transcription factors: Identifies genomic binding sites for STAT5 and other transcription factors activated by GM-CSF signaling.

  • Single-cell RNA-seq: Reveals cell-specific responses to GM-CSF in heterogeneous populations.

Functional Response Assays

  • Cell survival analysis: GM-CSF promotes survival of myeloid cells, assessable through Annexin V/PI staining and flow cytometry.

  • Cytokine production profiling: Multiplex analysis of cytokines produced by GM-CSF-stimulated cells reveals functional outcomes of receptor activation.

  • Migration assays: Transwell systems to assess how GM-CSF influences cellular motility and chemotaxis.

  • Phagocytosis and respiratory burst: Functional readouts of myeloid cell activation following GM-CSF stimulation.

The integration of these approaches has revealed that GM-CSF signaling in monocytes significantly enhances their inflammatory potential, particularly in autoimmune contexts. For example, GM-CSF priming increases anti-MPO-stimulated IL-1β production and promotes subsequent Th17 polarization, creating a feed-forward inflammatory loop in AAV .

How is recombinant mouse GM-CSF being utilized in COVID-19 and emerging infectious disease research?

While the search results don't specifically address COVID-19 research with mouse GM-CSF, this is an emerging area worthy of discussion based on the known functions of GM-CSF and its potential applications in infectious disease models.

Recombinant mouse GM-CSF has significant potential applications in COVID-19 and emerging infectious disease research, particularly in mouse models of infection and inflammatory response. The dual role of GM-CSF in both promoting protective immunity and potentially exacerbating harmful inflammation makes it a complex but important factor in infectious disease research.

In modeling COVID-19 pathogenesis:

  • GM-CSF contributes to hyperinflammation in severe COVID-19, with elevated levels detected in patients with severe disease. Mouse models using recombinant GM-CSF can help elucidate mechanisms of cytokine storm development.

  • The role of GM-CSF in lung myeloid cell recruitment and activation is particularly relevant to COVID-19 pathology. Administration of recombinant mouse GM-CSF at different timepoints in infection models could reveal temporal aspects of its contribution to disease progression.

In therapeutic development research:

  • GM-CSF blockade has been investigated in human COVID-19 as an anti-inflammatory strategy. Mouse models using anti-GM-CSF antibodies can help define optimal timing and targeting approaches.

  • Conversely, GM-CSF administration has been explored as a therapy for COVID-19-associated lymphopenia. Recombinant mouse GM-CSF could be used to investigate this approach in murine infection models.

Methodological considerations for infectious disease research:

  • When using recombinant mouse GM-CSF in infectious disease models, careful attention to dosing and timing relative to infection is critical, as effects may be beneficial or detrimental depending on context.

  • Integration with other cytokine measurements (especially IL-6, TNF-α, and type I interferons) provides a more comprehensive understanding of the inflammatory network involved in disease pathogenesis.

  • Combining GM-CSF manipulation with viral or bacterial challenge models requires rigorous biosafety protocols and specialized facilities, particularly for studies involving biosafety level 3 (BSL-3) pathogens.

What role does GM-CSF play in metabolic reprogramming of myeloid cells?

Recent research has revealed that GM-CSF significantly influences cellular metabolism in myeloid cells, which in turn affects their functional phenotype and inflammatory potential. This emerging area connects immunometabolism with GM-CSF biology:

Metabolic pathways regulated by GM-CSF:

  • Glycolysis enhancement: GM-CSF stimulation rapidly increases glycolytic flux in monocytes and macrophages, supporting their activation and inflammatory functions.

  • Mitochondrial metabolism: Beyond glycolysis, GM-CSF modulates oxidative phosphorylation and mitochondrial dynamics in myeloid cells.

  • Fatty acid metabolism: GM-CSF alters lipid metabolism, potentially influencing membrane composition and lipid mediator production.

Experimental approaches to study GM-CSF-induced metabolic changes:

  • Seahorse extracellular flux analysis: Measures oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) to quantify oxidative phosphorylation and glycolysis, respectively, in GM-CSF-stimulated cells.

  • Metabolite profiling: Mass spectrometry-based metabolomics reveals changes in metabolite levels and flux through various pathways following GM-CSF stimulation.

  • Stable isotope tracing: Using labeled glucose, glutamine, or fatty acids to track metabolic pathway activity and substrate utilization in response to GM-CSF.

  • Integration with transcriptomics: Connecting changes in metabolic enzyme expression with functional metabolic alterations provides mechanistic insights.

Functional consequences of metabolic reprogramming:

  • Enhanced IL-1β production: The increased IL-1β generation by GM-CSF-primed monocytes in response to anti-MPO stimulation may partially result from metabolic reprogramming, as glycolysis is required for optimal IL-1β processing and release.

  • Trained immunity development: GM-CSF may contribute to trained immunity through epigenetic modifications mediated by metabolic intermediates.

  • Antigen presentation capacity: Metabolic reprogramming influences antigen processing and presentation, potentially explaining GM-CSF's effects on adaptive immune responses.

This emerging area connects GM-CSF signaling with broader concepts in immunometabolism and offers new therapeutic targets at the intersection of metabolism and inflammation.

How can recombinant mouse GM-CSF be utilized in cancer immunotherapy research?

Recombinant mouse GM-CSF has multiple applications in cancer immunotherapy research, serving both as a tool to understand fundamental biology and as a component of experimental therapeutics:

Fundamental research applications:

  • Dendritic cell generation: Recombinant mouse GM-CSF is essential for generating bone marrow-derived dendritic cells in vitro, which can be loaded with tumor antigens and used in vaccination studies.

  • Macrophage polarization: GM-CSF influences macrophage phenotype and function, with implications for tumor-associated macrophage biology.

  • Myeloid-derived suppressor cell (MDSC) development: GM-CSF can promote MDSC expansion, providing a system to study these immunosuppressive cells.

Therapeutic strategy development:

  • GM-CSF-secreting tumor vaccines: Mouse tumor cells engineered to secrete GM-CSF have been used to develop GVAX-type vaccines, which enhance immune responses against tumor antigens.

  • Combination therapy models: Recombinant GM-CSF can be combined with checkpoint inhibitors in mouse models to investigate synergistic effects.

  • Adoptive cell therapy enhancement: GM-CSF can be used to condition myeloid cells for adoptive transfer or to support in vivo expansion of transferred immune cells.

Technical considerations for cancer research:

  • Dose optimization: Both dose and timing of GM-CSF administration are critical, as high levels may promote MDSCs while lower levels may enhance anti-tumor immunity.

  • Strain selection: Different mouse strains show variable responses to GM-CSF and cancer immunotherapies. C57BL/6 mice are commonly used but may not always represent human responses.

  • Tumor model selection: The effects of GM-CSF manipulation vary depending on tumor type, immunogenicity, and microenvironment.

  • Comparison with human systems: Remember that mouse and human GM-CSF have no cross-reactivity , necessitating careful consideration when translating findings to human applications.

This research area continues to evolve as our understanding of myeloid cell complexity in the tumor microenvironment deepens, with GM-CSF playing multifaceted roles that vary by context and timing.

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.