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

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Lyophilized from a 0.2µm filtered PBS, pH 7.4.
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. Please reconstitute protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers could use it as a reference.
Shelf Life
The shelf life is influenced by various factors such as storage conditions, buffer ingredients, storage temperature, and the inherent 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 uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
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
14.1 kDa
Protein Length
Full Length of Mature Protein
Purity
>98% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that plays a crucial role in the growth and differentiation of hematopoietic precursor cells from various lineages, including granulocytes, macrophages, eosinophils, and erythrocytes.
Gene References Into Functions
  1. Studies have shown that the microbiota enhances respiratory defenses through GM-CSF signaling, stimulating pathogen killing and clearance by alveolar macrophages. PMID: 29142211
  2. Research suggests that GM-CSF contributes to chronic tissue damage and disability in experimental autoimmune encephalomyelitis through diverse pathways, although it is not essential during early lesion formation or the onset of neurological deficits. PMID: 29288202
  3. The release of IL-33 and GM-CSF from epithelial cells triggers the activation of p65 and the p38-MK2/3 signaling module in dendritic cells, leading to Th2 polarization and ultimately, allergic inflammation. PMID: 29288203
  4. Findings indicate that T cell production of GM-CSF is essential for controlling M. tuberculosis infection in the absence of other GM-CSF sources. Multiple T cell subsets produce GM-CSF in the lung during infection, and GM-CSF directly acts on infected macrophages via a PPARgamma-dependent pathway to limit bacterial growth. PMID: 29066547
  5. In conclusion, studies have confirmed the pathogenic role of GM-CSF in colitis-associated colorectal cancer development. GM-CSF promotes a 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 serve as a prognostic biomarker for lung and esophageal cancer. PMID: 27835886
  7. This study shows that epithelial-derived GM-CSF is a critical early signal during allergic sensitization to cockroach allergen. PMID: 27731325
  8. The impaired macrophage functions in leukemic mice were significantly improved by IL-3 and GM-CSF treatment, suggesting the therapeutic potential of these 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. Both T-GM-CSF and -IL-3 significantly and reciprocally inhibited 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 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 essential for the normal balance of leukocyte subsets, including granulocytes, B cells, and naïve 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 cells, respectively, via its receptors, consequently impairing MDSC induction, 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 the 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, causing 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, but 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 hematoma. 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 exhibited 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, supporting 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 the 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 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. Findings suggest that over-production of GM-CSF by T cells could be a pathogenic factor in many histiocytic disorders. PMID: 24183847
  48. 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 protein and what are its main biological functions?

Recombinant mouse GM-CSF (Granulocyte-Macrophage Colony-Stimulating Factor), also known as Csf2, is a cytokine initially characterized for its ability to support the in vitro colony formation of granulocyte-macrophage progenitors. The E. coli-derived recombinant protein typically consists of amino acids Ala18-Lys141, with an N-terminal Met .

Biologically, mouse GM-CSF functions as:

  • A growth factor for erythroid, megakaryocyte, and eosinophil progenitors

  • A survival factor for mature hematopoietic cells

  • An activator of effector functions in granulocytes, monocytes/macrophages, and eosinophils

  • A promoter of Th1-biased immune responses, angiogenesis, and allergic inflammation

  • A contributor to autoimmunity development

GM-CSF exerts its biological effects through a heterodimeric receptor complex composed of GM-CSF Rα/CD116 (the specific binding subunit) and the common β chain (CD131), which is also a component of the IL-3 and IL-5 receptors .

What are the structural characteristics of recombinant mouse GM-CSF?

Recombinant mouse GM-CSF has a molecular weight of approximately 14.3 kDa as analyzed by SEC-MALS (Size Exclusion Chromatography with Multi-Angle Light Scattering), suggesting that the protein exists as a monomer . Under reducing SDS-PAGE conditions, it appears as a single band at approximately 14 kDa when visualized by silver staining . In its native form, mouse GM-CSF typically runs as a 15.5-19 kDa band in both reducing and non-reducing SDS-PAGE analysis .

Structurally, mouse GM-CSF contains 124 amino acid residues and, similar to IL-3 and IL-5, has a core of four bundled alpha-helices . Mature mouse GM-CSF shares 49%-54% amino acid sequence identity with canine, feline, human, and porcine GM-CSF, and 69% with rat GM-CSF .

What are the recommended storage and reconstitution protocols for recombinant mouse GM-CSF?

For optimal activity preservation, follow these guidelines:

Storage:

  • Store at -80°C in a manual defrost freezer

  • Avoid repeated freeze-thaw cycles

  • Upon initial thawing, aliquot into polypropylene microtubes before refreezing

Reconstitution by formulation type:

  • For lyophilized product with carrier protein (e.g., 415-ML):

    • Reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin

  • For carrier-free lyophilized product (e.g., 415-ML/CF):

    • Reconstitute at 100 μg/mL in sterile PBS

  • For dilution and long-term storage:

    • For in vitro biological assays: Use carrier protein concentrations of 0.5-1 mg/mL

    • For ELISA standards: Use carrier protein concentrations of 5-10 mg/mL

    • Never dilute below 5 μg/mL for long-term storage

When should carrier-free versions of recombinant mouse GM-CSF be used?

Carrier-free (CF) formulations of recombinant mouse GM-CSF do not contain Bovine Serum Albumin (BSA) or other carrier proteins. They are typically lyophilized from 0.2 μm filtered solutions in PBS .

These formulations are specifically recommended for applications where the presence of BSA could interfere, such as:

  • Studies requiring pure protein interactions without carrier protein interference

  • Experimental systems sensitive to bovine proteins

  • Conjugation or labeling applications where carrier proteins might compete for reactive sites

  • Mass spectrometry or other analytical techniques where carrier proteins could confound results

Researchers should be aware that carrier proteins may have undesired influence on experimental results due to potential toxicity, high endotoxin levels, or possible blocking activity .

How is the biological activity of recombinant mouse GM-CSF measured?

The biological activity of recombinant mouse GM-CSF can be assessed through:

  • Cell proliferation assays:

    • The standard assay uses the DA3 mouse myeloma cell line

    • The effective dose (ED50) typically ranges from 5-30 pg/mL

    • This remarkably low ED50 demonstrates the high potency of the protein

  • Colony formation assays:

    • Using bone marrow progenitor cells to measure colony-forming units of granulocyte-macrophage lineage

  • Functional activation assays:

    • Measuring respiratory burst in neutrophils or macrophages

    • Cytokine production by stimulated cells

    • Phagocytic activity enhancement

  • Receptor binding assays:

    • Using cells expressing mouse GM-CSF receptor complex

For quality control purposes, recombinant mouse GM-CSF preparations should have endotoxin levels ≤ 0.1 ng per μg of protein, as measured by chromogenic LAL assay, and ≥ 95% purity as determined by SDS-PAGE and absorbance measurements .

How does GM-CSF contribute to autoimmune and inflammatory diseases in mouse models?

GM-CSF has been implicated in several autoimmune and inflammatory conditions through multiple mechanisms:

Experimental Autoimmune Prostatitis (EAP):

  • Elevated GM-CSF mRNA levels have been detected in prostate tissue from EAP mice

  • GM-CSF may contribute to both inflammatory responses in prostate tissue and pain development through neuronal mechanisms

  • GM-CSF knockout mice show decreased symptom severity in EAP models

Atherosclerosis:

  • In LDL-receptor-deficient (Ldlr-/-) mice, GM-CSF promotes advanced plaque progression

  • The mechanism involves increasing macrophage apoptosis susceptibility through:

    • GM-CSF-mediated production of IL-23

    • IL-23-induced proteasomal degradation of the cell-survival protein Bcl-2

    • Increased oxidative stress

  • GM-CSF-deficient mice crossed with Ldlr-/- mice show substantial decreases in lesional macrophage apoptosis and plaque necrosis

These findings highlight GM-CSF as a potential therapeutic target in various inflammatory and autoimmune conditions, with its specific role varying depending on the disease context and affected tissues.

What experimental approaches can be used to study GM-CSF function in disease models?

Researchers can employ several experimental approaches to investigate GM-CSF function:

Genetic Models:

  • GM-CSF knockout mice (Csf2-/-) - Complete absence of GM-CSF allows assessment of its necessity in disease development

  • Cell-specific conditional knockouts - Targeting GM-CSF or its receptor in specific cell populations

  • Transgenic overexpression - To study effects of increased GM-CSF levels

Pharmacological Approaches:

  • Neutralizing antibodies - For temporal and dose-dependent inhibition

  • Recombinant GM-CSF administration - To supplement or rescue phenotypes

  • Receptor antagonists - To block signaling without affecting protein levels

Analytical Methods:

  • mRNA quantification - Using qRT-PCR to measure GM-CSF expression in tissue samples

  • Protein detection - ELISA, Western blotting, or immunohistochemistry

  • Functional assays - As described in question 1.5

  • Signaling pathway analysis - Phospho-flow cytometry, Western blotting for active signaling molecules

Experimental Design Considerations:

  • Include appropriate controls (isotype controls, vehicle treatments)

  • Validate findings using multiple approaches

  • Consider potential compensatory mechanisms in knockout models

  • Account for strain-specific differences in GM-CSF responses

  • Remember the species-specific nature of GM-CSF activity - mouse GM-CSF shows limited cross-reactivity with rat cells

How does GM-CSF influence macrophage function and apoptosis in disease models?

GM-CSF exerts significant effects on macrophage biology that impact disease pathogenesis:

Macrophage Differentiation and Polarization:

  • Promotes differentiation of monocytes into inflammatory macrophages

  • Influences the balance between M1 (pro-inflammatory) and M2 (tissue repair) phenotypes

  • Affects expression of scavenger receptors and pattern recognition receptors

Apoptosis Regulation in Atherosclerosis:

  • Increases macrophage susceptibility to apoptosis in advanced atherosclerotic plaques

  • The mechanism involves:

    • GM-CSF-induced IL-23 production

    • IL-23-mediated downregulation of the anti-apoptotic protein Bcl-2

    • Enhanced sensitivity to atherosclerosis-relevant pro-apoptotic factors (7-ketocholesterol, oxidized-LDL)

  • GM-CSF-deficient (Csf2-/-Ldlr-/-) mice show reduced lesional macrophage apoptosis and plaque necrosis

Inflammatory Activation:

  • Enhances production of pro-inflammatory cytokines and chemokines

  • Increases reactive oxygen species (ROS) generation

  • Augments phagocytic activity and antigen presentation

Tissue-Specific Effects:

  • In prostate tissue (EAP model), affects inflammatory cell recruitment and activation

  • In arterial plaques, contributes to advanced lesion progression and vulnerability

  • Effects may vary depending on the tissue microenvironment and disease stage

These diverse effects make GM-CSF a potent regulator of macrophage function in multiple disease contexts, with potential for therapeutic targeting.

What role does GM-CSF play in the experimental autoimmune prostatitis (EAP) mouse model?

In the experimental autoimmune prostatitis (EAP) mouse model, GM-CSF has been identified as a significant contributor to disease pathogenesis:

Expression Pattern:

  • Elevated GM-CSF mRNA levels are detected in prostate tissue from EAP mice

  • This mirrors findings in expressed prostatic secretions from human chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) patients

Receptor Distribution:

  • GM-CSF receptors are expressed in both mouse prostate tissue and dorsal root ganglia

  • This dual expression suggests potential roles in both local inflammation and pain signaling

Functional Impact:

  • GM-CSF knockout mice (Csf2-/-) show reduced symptom severity when EAP is induced, including:

    • Decreased behavioral indicators of pain

    • Reduced prostatic inflammation scores

    • Lower expression of nociceptive and inflammatory markers

Mechanistic Insights:

  • May contribute to immune cell recruitment and activation in prostate tissue

  • Potentially modulates nociceptive pathways, given receptor expression in dorsal root ganglia

  • Could represent a link between prostatic inflammation and pelvic pain in this model

These findings suggest that GM-CSF plays dual roles in EAP pathogenesis - contributing to both inflammatory responses in prostate tissue and potentially to pain development through neuronal mechanisms, making it a promising therapeutic target for CP/CPPS.

What are the signaling pathways downstream of GM-CSF receptor activation in mice?

GM-CSF receptor signaling in mice involves a complex network of pathways following binding to its heterodimeric receptor:

Receptor Complex:

  • GM-CSF Rα/CD116: The specific binding subunit

  • Common β chain (CD131): The signaling subunit shared with IL-3 and IL-5 receptors

Major Signaling Cascades:

  • JAK-STAT Pathway:

    • JAK2 associates with the β chain and becomes activated

    • Leads to phosphorylation and activation of STAT5, and to a lesser extent STAT3

    • Activated STATs translocate to the nucleus and regulate gene expression

    • Critical for proliferation and survival signals

  • PI3K/Akt Pathway:

    • Important for cell survival signals

    • Regulates anti-apoptotic proteins including Bcl-2 family members

    • Disruption in this pathway can enhance macrophage apoptosis susceptibility as observed in atherosclerosis models

  • MAPK Pathways:

    • Activation of ERK1/2, p38 MAPK, and JNK

    • Contributes to proliferation, differentiation, and inflammatory responses

    • Plays a role in cytokine production by activated cells

  • NF-κB Pathway:

    • Important for inflammatory gene expression

    • Regulates production of cytokines, including IL-23

    • IL-23 has been implicated in GM-CSF-mediated effects in atherosclerosis

Pathway Crosstalk:

  • These pathways interact extensively and show context-dependent activation

  • The balance between these signals determines cellular outcomes including survival, proliferation, differentiation, and activation

Understanding these signaling mechanisms provides potential points for therapeutic intervention in GM-CSF-mediated pathologies.

How does recombinant mouse GM-CSF interact with other cytokines in inflammatory networks?

Recombinant mouse GM-CSF functions within a complex cytokine network during inflammatory responses:

GM-CSF and the IL-23/IL-17 Axis:

  • GM-CSF stimulates production of IL-23, which mediates some of GM-CSF's pro-inflammatory and pro-apoptotic effects in atherosclerosis models

  • In turn, IL-23 can stimulate IL-17 production by T cells

  • IL-17 may further enhance GM-CSF production, creating a positive feedback loop

Interactions with Pro-inflammatory Cytokines:

  • Synergizes with TNF-α to enhance inflammatory activation of macrophages

  • Amplifies IL-1β production and signaling in myeloid cells

  • Interferon-γ can enhance GM-CSF receptor expression and signaling

Regulation by Anti-inflammatory Cytokines:

  • IL-10 can suppress GM-CSF production and some downstream effects

  • TGF-β may modulate GM-CSF-induced myeloid cell differentiation

  • IL-4 can alter macrophage responses to GM-CSF

Methodological Approaches for Studying Cytokine Interactions:

  • Multi-parameter flow cytometry to analyze cell-specific responses

  • Cytokine co-stimulation experiments in vitro

  • Multiplex cytokine profiling in disease models

  • Combined cytokine blockade or knockout models

  • Transcriptomic analysis to identify cytokine networks

What are the experimental challenges when working with recombinant mouse GM-CSF in research?

Researchers face several technical and interpretative challenges when studying GM-CSF activity:

Technical Challenges:

  • Protein Stability Issues:

    • Limited stability in solution

    • Requires carrier proteins (e.g., BSA) for stabilization

    • Needs storage at -80°C and avoidance of freeze-thaw cycles

    • Concentration should not be diluted below 5 μg/mL for long-term storage

  • Detection Sensitivity Requirements:

    • Biological activity is potent at very low concentrations (ED50 of 5-30 pg/mL)

    • Requires highly sensitive detection methods

    • Standard curves must cover wide concentration ranges

  • Formulation Considerations:

    • Carrier proteins may cause experimental interference

    • Need to select between standard and carrier-free formulations based on application

    • Endotoxin contamination must be minimal (≤0.1 ng per μg)

Experimental Design Challenges:

  • Biological Complexity:

    • Overlapping functions with IL-3 and IL-5 (shared β chain receptor)

    • Context-dependent activity across different cell types and tissues

    • Species specificity limiting translational applications

  • Interpretation Difficulties:

    • Compensatory mechanisms in knockout models

    • Distinguishing direct vs. indirect effects through cytokine networks

    • Variations in receptor expression across cell populations

Methodological Solutions:

ChallengePotential Solutions
Protein stabilityUse fresh aliquots; include carrier protein; verify activity regularly
Assay sensitivityUse low-passage sensitive cell lines; optimize detection methods
Compensatory mechanismsInclude multiple time points; combine genetic and antibody approaches
Species specificityUse species-matched systems; caution in translational interpretations
Context dependencyTest multiple cell types and physiological conditions

These considerations highlight the importance of rigorous experimental design and appropriate controls when working with recombinant mouse GM-CSF in research applications.

How can GM-CSF knockout mice be used to elucidate cytokine function in disease models?

GM-CSF knockout mice (Csf2-/-) serve as valuable tools for investigating this cytokine's role in various disease processes:

Experimental Design Strategies:

  • Disease Model Crosses:

    • GM-CSF knockout mice can be crossed with disease-specific models (e.g., Ldlr-/- for atherosclerosis)

    • This approach allows assessment of GM-CSF's role in specific pathological contexts

  • Induced Disease Models:

    • Direct application of disease induction in GM-CSF knockout background (e.g., EAP induction)

    • Comparing disease progression between knockout and wildtype mice

  • Bone Marrow Chimeras:

    • Transplanting GM-CSF-deficient bone marrow into wildtype recipients (or vice versa)

    • Helps distinguish between hematopoietic and non-hematopoietic sources of GM-CSF

Parameter Assessment:

  • Clinical Evaluation:

    • Disease-specific clinical scores

    • Physiological parameters (weight, temperature, behavior)

    • Functional assessments (e.g., pain sensitivity in EAP models)

  • Cellular Analysis:

    • Flow cytometry to assess immune cell populations

    • Histopathological evaluation of affected tissues

    • Cell subset function and activation status

  • Molecular Assessment:

    • Cytokine and inflammatory mediator profiling

    • Gene expression analysis of disease-relevant pathways

    • Signaling molecule activation status

Mechanistic Investigations:

  • Differences in disease outcomes can be analyzed to elucidate downstream mechanisms

  • Example: Decreased macrophage apoptosis in atherosclerotic plaques of Csf2-/-Ldlr-/- mice revealed GM-CSF's role in promoting cell death through IL-23 and Bcl-2 regulation

Validation Approaches:

  • Rescue experiments with recombinant GM-CSF administration

  • Antibody-mediated GM-CSF neutralization in wildtype mice

  • Cell-specific conditional knockout models for refined mechanistic insights

When interpreting results from knockout models, researchers should consider potential compensatory mechanisms and developmental effects that might influence outcomes independently of GM-CSF's direct role in pathogenesis.

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.