Recombinant Human Granulocyte-Macrophage Colony Stimulating Factor (CSF2), partial (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

rhGM-CSF modulates immune responses through:

  • Stimulating hematopoiesis: Accelerates recovery of neutrophils, monocytes, and eosinophils post-chemotherapy .

  • Activating immune cells: Enhances phagocytosis, superoxide production, and cytotoxic activity in neutrophils and eosinophils .

  • Signaling pathways: Triggers STAT5 phosphorylation for proliferation and STAT3 for macrophage activation, inducing zinc deprivation and reactive oxygen species to combat fungi .

Hematological Malignancies and Infections

  • Refractory invasive fungal diseases (IFDs): Adjunctive therapy with sargramostim (a yeast-derived rhGM-CSF) achieved an 82% response rate in pediatric patients with hematological malignancies .

  • Febrile neutropenia: Reduced hospitalization duration (252 vs. 354 days) and antibiotic usage in a randomized trial .

Sepsis and Pulmonary Dysfunction

  • Low-dose GM-CSF (3 μg/kg/day) improved PaO₂/FiO₂ ratios and restored leukocyte phagocytic function in sepsis patients without worsening organ dysfunction .

Burns and Wound Healing

  • rhGM-CSF gel combined with medical collagen sponges accelerated healing of deep second-degree burns in infants, outperforming EGF/FGF therapies .

Autoimmune Diseases

  • Dual role: While GM-CSF promotes Th1 responses and inflammation in conditions like rheumatoid arthritis , it also induces tolerogenic dendritic cells (CD8a⁻) to suppress autoimmune reactions (e.g., myasthenia gravis) .

Production and Quality Standards

rhGM-CSF is produced via recombinant DNA technology in:

  • Expression systems: E. coli (non-glycosylated) or yeast (glycosylated) .

  • GMP compliance: Manufactured in ISO-certified facilities with lot-to-lot consistency and bioactivity validation .

ParameterSpecificationSource
Purity>95% via SDS-PAGE and HPLC
Endotoxin Content<1 EU/μg
Storage-20°C for lyophilized form; avoid repeated freeze-thaw

Research Gaps and Future Directions

  • Tumor microenvironment: GM-CSF’s dual role in promoting/suppressing tumor growth warrants further investigation for cancer immunotherapy .

  • Combination therapies: Synergistic effects with antifungals or antibiotics in refractory infections require exploration .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
Typically, we can ship your order within 5-10 business days after receiving it. However, delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery information.
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 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 several 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. The shelf life of 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
Colony stimulating factor 2 (granulocyte-macrophage); Colony Stimulating Factor 2; Colony stimulating factor; Colony-stimulating factor; CSF 2; CSF; CSF2; CSF2_HUMAN; GM-CSF; GMCSF; Granulocyte Macrophage Colony Stimulating Factor; Granulocyte-macrophage colony-stimulating factor; MGC131935; MGC138897; MGI1GM; Molgramostin; Pluripoietin-a; Sargramostim
Datasheet & Coa
Please contact us to get it.
Expression Region
18-144aa
Mol. Weight
14.5 kDa
Protein Length
Partial
Purity
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Immunology
Source
E.Coli
Species
Homo sapiens (Human)
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. GM-CSF can be induced by *M. pneumoniae* infection *in vivo* and *in vitro*. Children with high levels of GM-CSF experienced longer durations of fever. GM-CSF likely plays a crucial role in neutrophil inflammation during *M. pneumoniae* infection. PMID: 30007865
  2. Data demonstrate that CD4 T cells from synovial fluid are enriched for granulocyte-macrophage colony-stimulating factor (GM-CSF) production, suggesting that the GM-CSF program may be a primary pathogenic process. PMID: 29142230
  3. GM-CSF bioavailability plays a critical role in maintaining intestinal homeostasis. Decreased bioavailability, coupled with genetic risk markers and/or smoking, results in aggressive ileal Crohn's disease behavior. PMID: 29434451
  4. Data indicate that GIF (ORF117) serves as a competitive decoy receptor by leveraging binding hotspots underlying the cognate receptor interactions of granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2). PMID: 27819269
  5. Translated to human disease, these findings suggest that vaccine-mediated expansion of GM-CSF-producing T cells could be an effective prophylactic or therapeutic strategy for tuberculosis. PMID: 29233902
  6. In a subset of patients who received GM-CSF, circulating myeloid-derived suppressor cells (MDSC) and anti-GM-CSF-neutralizing antibodies (Nabs) were also modulated. The majority of patients developed anti-GM-CSF Nabs, which correlated with improved RFS and OS. PMID: 28536308
  7. This study demonstrates that cortisol inhibits CSF2 via DNA methylation and inhibits invasion in first-trimester trophoblast cells. PMID: 28846166
  8. Oral and periodontal innate immunity is affected by HIV viremia and ART. GCF IL-8, G-CSF, as well as serum IL-8, MCP-1, and GM-CSF may be useful biomarkers for the detection of disease presence and/or its severity due to HIV infection and ART use. PMID: 28043030
  9. This review summarizes numerous findings to provide the currently available information regarding the anticancer immune response of GM-CSG. [review] PMID: 27364892
  10. Data suggest that intratumoral GM-CSF expression, as a potentially independent prognostic biomarker for recurrence, might improve conventional clinical and pathologic analysis to refine outcome prediction for clinically localized clear-cell renal cell carcinoma (ccRCC) patients after surgery. PMID: 27016418
  11. High GM-CSF expression is associated with breast cancer. PMID: 28754674
  12. In gastric cancer (GC), tumor-derived GM-CSF activated neutrophils and induced neutrophil PD-L1 expression via the Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. The activated PD-L1(+) neutrophils effectively suppressed normal T-cell immunity *in vitro* and contributed to the growth and progression of human GC *in vivo*. PMID: 28274999
  13. The IL-3/GM-CSF effect on the myofibroblastic differentiation of human adipose-derived stromal cells (hASCs) is similar to that observed in human dermal fibroblasts (HDFs). PMID: 28377320
  14. Obesity alters lung neutrophil infiltration to enhance breast cancer metastasis through IL5 and GM-CSF. PMID: 28737771
  15. Data suggest that the methylotrophic yeast *Pichia pastoris* is an effective recombinant host for heterologous granulocyte-macrophage colony-stimulating factor (rhGM-CSF) production. PMID: 28376863
  16. Letter: Knockdown of either filaggrin or loricrin increases the production of interleukin (IL)-1alpha, IL-8, IL-18, and granulocyte macrophage colony-stimulating factor in stratified human keratinocytes. PMID: 26381575
  17. Impaired RASGRF1/ERK-mediated GM-CSF response characterizes CARD9 deficiency in French-Canadians. PMID: 26521038
  18. Honokiol may possess potential anti-inflammatory effects and inhibits TNF-alpha-induced IL-1beta, GM-CSF, and IL-8 production in PBMCs from rheumatoid arthritis patients. PMID: 26339358
  19. The canonical NFkappaB signaling in fibroblasts, but not in tumor cells, was shown to be responsible for the induced and constitutive CSF2 expression. PMID: 25919140
  20. GM-CSF primes IL-13 production by macrophages via PAR-2. PMID: 25633855
  21. Analysis of an IL2RA polymorphism that controls GM-CSF production in human TH cells. PMID: 25278028
  22. NFKBIZ gene knockdown in bronchial epithelial cells suppresses the release of IL-1b-induced IL-6 and GMCSF. PMID: 25629767
  23. CFH Y402H polymorphism is associated with elevated vitreal GM-CSF and choroidal macrophages in the postmortem human eye. PMID: 25814824
  24. Influenza infection stimulates the secretion of IL-8 and GM-CSF by alveolar epithelial cells. PMID: 26033355
  25. Periodic fever, aphthous stomatitis, pharyngitis, and cervical adenopathy syndrome febrile episodes are characterized by activation of GM-CSF and IL-8 with Th1 skewing. PMID: 24670131
  26. Activation of STAT3 induces the expression of GM-CSFRa that protects CLL cells from apoptosis, suggesting that inhibition of STAT3 or GM-CSFRa may benefit patients with chronic lymphocytic leukemia. PMID: 24836891
  27. G-CSF- or GM-CSF-secreting cancers, albeit not very common, are, however, among the most rapidly advancing ones due to a cytokine-mediated immune suppression and angiogenesis. [Review] PMID: 24692240
  28. Treatment with GM-CSF increased the expression of vimentin and ZEB1, similar to TGF-b1 treatment, and decreased the expression of E-cadherin. These results suggest that GM-CSF could induce EMT in human HERS/ERM cells. PMID: 24258001
  29. GM-CSF polymorphism may be involved in the pathogenesis of atopic dermatitis. PMID: 24117406
  30. The association of GM-CSF expression with systemic inflammation highlights the potential role of Th17-related cytokines in the pathology of juvenile idiopathic arthritis. PMID: 24692225
  31. M-CSF induces macrophage transformation by upregulating c-Jun with a certain synergy of NFkappaB. PMID: 24100343
  32. The detection of basal levels of IgM rheumatoid factor, IgM, and certain cytokines can be useful in prognosticating the effectiveness of rituximab therapy for rheumatoid arthritis. PMID: 25080789
  33. Cytokines and signaling molecules predict clinical outcomes in sepsis. PMID: 24244449
  34. Granulocyte colony-stimulating factor produces long-term changes in gene and microRNA expression profiles in CD34+ cells from healthy donors. PMID: 24056818
  35. Mesenchymal-like breast cancer cells activate macrophages to a tumor-associated macrophage-like phenotype by GM-CSF. PMID: 24823638
  36. The comparison between the cytokine profiles of ET and PV patients showed a statistically significant increase of IL-4, IL-8, GM-CSF, interferon-gamma, MCP-1, PDGF-BB, and VEGF in the ET group. PMID: 24463275
  37. Data indicate that differentially expressed IL-17, IL-22, and IL-23 levels are associated with K-ras in a stage-specific fashion along colorectal cancer progression, and an association was established between mutant K-ras and GM-CSF and IFN-gamma. PMID: 24040001
  38. In the type 2 diabetes mellitus group, significantly higher IL-4 levels were observed on the first and third post-dengue virus infection days, while higher IL-10 and granulocyte-macrophage colony-stimulating factor were detected on the third post-infection day. PMID: 24078930
  39. Loss of BMPR2 induced prolonged phospho-p38 MAPK in response to TNF, and this increased GADD34-PP1 phosphatase activity, dephosphorylating eukaryotic translation initiation factor (eIF2alpha), and derepressing GM-CSF mRNA translation. PMID: 24446489
  40. Given that GM-CSF plays an important role during pregnancy, the reduced levels of GM-CSF in the cervico-vaginal fluid of human papillomavirus(+) women may contribute to explaining the reduced live birth rate. PMID: 23920355
  41. SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells. PMID: 24211252
  42. Describes a GM-CSF ELISA for routine clinical diagnosis of autoimmune pulmonary alveolar proteinosis. PMID: 24275678
  43. The data presented suggest that -677C/A in the promoter region of the GM-CSF gene does not play a role in acne pathogenesis. PMID: 23069312
  44. CBL linker and RING finger mutants lead to enhanced GM-CSF signaling due to elevated kinase expression, which can be blocked using small molecule inhibitors targeting specific downstream pathways. PMID: 23696637
  45. Impairment of NFATC2-calcineurin interaction promotes megakaryocyte proliferation through up-regulation of CSF2 transcription. PMID: 22911897
  46. Overexpression of a nonphosphorylatable MT1-MMP mutant (Y573F) abrogates CSF-2 and CSF-3 transcriptional increases. PMID: 23548906
  47. GM-CSF contributes to aortic aneurysms resulting from SMAD3 deficiency. PMID: 23585475
  48. Within pediatric Crohn disease, neutrophil GM-CSF-dependent STAT5 activation and bacterial killing were reduced as GM-CSF autoantibodies increased. PMID: 23600834
  49. This study demonstrated that GMCSF is downregulated in the brain of patients with Alzheimer's disease. PMID: 22430742
  50. IL-27 suppresses GM-CSF production under nonpolarizing and T helper (Th)1 cell conditions, but not T helper (Th)17 cell conditions, in both murine and human cultures. PMID: 22837488

Show More

Hide All

Database Links

HGNC: 2434

OMIM: 138960

KEGG: hsa:1437

STRING: 9606.ENSP00000296871

UniGene: Hs.1349

Protein Families
GM-CSF family
Subcellular Location
Secreted.

Q&A

What is Recombinant Human GM-CSF and how does it differ from endogenous GM-CSF?

Recombinant Human GM-CSF (rhu GM-CSF) is a laboratory-produced version of the naturally occurring cytokine that was identified in the 1960s as a myeloid growth factor, purified in the 1970s, molecularly-cloned in the 1980s, and clinically developed in the 1990s . The recombinant form mimics the structure and function of endogenous GM-CSF but is produced through molecular cloning techniques in various expression systems including E. coli, yeast, or mammalian cells.

Structurally, E. coli-derived human GM-CSF protein typically encompasses amino acids Ala18-Glu144 of the native sequence . Unlike endogenous GM-CSF, recombinant versions may exhibit differences in glycosylation patterns depending on the expression system used, which can influence their pharmacokinetics, biological activity, and safety profiles . For research applications, recombinant GM-CSF is manufactured under controlled conditions, with GMP-grade productions adhering to strict quality standards for potential clinical applications .

The specific activity of recombinant human GM-CSF typically exceeds 1.0 × 10^7 IU/mg, calibrated against the human GM-CSF WHO International Standard (NIBSC code: 88/646) . In functional assays, it demonstrates potent biological activity, with an ED50 of 6-30 pg/mL in TF-1 cell proliferation assays .

What are the key biological activities of GM-CSF at the cellular level?

GM-CSF exerts multiple biological effects at the cellular level that extend well beyond its originally recognized role in myeloid cell development. These activities include:

  • Mitochondrial Function Enhancement: GM-CSF positively affects mitochondrial function and cellular metabolism in target cells, particularly within the mononuclear phagocyte system .

  • Phagocytosis and Efferocytosis Augmentation: It significantly enhances the phagocytic capacity of myeloid cells, improving clearance of pathogens and dying cells (efferocytosis) .

  • Immune Cell Modulation: GM-CSF regulates various immune cell functions, including antigen presentation, cytokine production, and inflammatory responses .

  • Trophoblast Differentiation: In placental development, GM-CSF influences trophoblast differentiation and functional maturation, impacting placental structure and potentially fetal growth .

  • Cell Survival Promotion: The cytokine supports survival of target cells, particularly in non-lymphoid tissues where it contributes to DC homeostasis .

  • Multilineage Effects: Research demonstrates that GM-CSF acts as a multilinear growth factor, affecting both erythroid precursors and granulocytes in bone marrow cells .

These diverse activities position GM-CSF as a critical regulator of immune function with therapeutic potential across multiple disease contexts where immune cell dysfunction occurs.

How does GM-CSF signaling work in target cells?

GM-CSF signaling involves a complex cascade that initiates with ligand binding to its heterodimeric receptor composed of an alpha subunit (CSF2RA/CD116) that confers specificity and a beta subunit (CSF2RB/CD131) shared with IL-3 and IL-5 receptors. The signaling mechanism involves:

  • Receptor Expression Patterns: The GM-CSF receptor is predominantly expressed on myeloid lineage cells. In placental tissues, CSF2RA is present in cytotrophoblasts and invading extravillous trophoblasts but shows weaker expression in syncytial trophoblasts .

  • Activation of Downstream Pathways: Upon ligand binding, the receptor activates multiple intracellular signaling cascades, including JAK-STAT, MAPK/ERK, and PI3K/Akt pathways, which mediate the diverse biological effects of GM-CSF.

  • Metabolic Reprogramming: GM-CSF signaling supports and restores the metabolic capacity of mononuclear phagocytes, which is fundamental to their functional capabilities .

  • Transcriptional Regulation: GM-CSF signaling influences the expression of multiple genes, including those controlling trophoblast differentiation (Ascl2, Tcfeb, Itgav, and Socs3) as demonstrated in placental development studies .

  • Paracrine Networks: GM-CSF participates in complex cytokine networks involving IL-1, TNF, IL-23, and IL-6, creating positive feedback loops that can sustain inflammatory responses in certain pathological conditions .

Understanding these signaling mechanisms is crucial for developing targeted therapies and interpreting experimental results in GM-CSF research.

How do different expression systems affect the glycosylation and activity of recombinant GM-CSF?

The expression system used to produce recombinant GM-CSF significantly impacts its glycosylation pattern and consequently its biological properties. Research has identified three distinct rhu GM-CSF formulations with varying glycosylation profiles based on their production platforms:

  • E. coli-derived GM-CSF: Produced in prokaryotic systems, this form lacks glycosylation entirely. The E. coli-derived human GM-CSF protein typically spans amino acids Ala18-Glu144 . In MALDI-TOF analysis, it shows a major peak corresponding to the calculated molecular mass of 14,478 Da, with potential matrix-associated artifacts .

  • Yeast-derived GM-CSF (ryGM-CSF): This form contains hyperglycosylation patterns characteristic of yeast expression systems. Interestingly, experimental evidence indicates that ryGM-CSF may exhibit enhanced potency compared to E. coli-derived forms, with a 2-fold proliferative effect on TF-1 cells achieved at only 0.064 ng/mL compared to 0.11 ng/mL for the E. coli counterpart .

  • Mammalian cell-derived GM-CSF: This form most closely mimics the natural glycosylation pattern of human GM-CSF.

The glycosylation differences between these formulations influence:

  • Pharmacokinetics: Affecting half-life and distribution in vivo

  • Biological Activity: Impacting receptor binding and signaling potency

  • Immunogenicity: Potentially affecting recognition by the host immune system

  • Stability: Influencing the protein's resistance to degradation

Researchers should carefully consider these variations when selecting a recombinant GM-CSF formulation for their specific experimental needs, as they may significantly impact experimental outcomes and interpretation of results.

What are the mechanisms behind GM-CSF's effects on mononuclear phagocyte metabolism and function?

GM-CSF exerts profound effects on mononuclear phagocyte metabolism and function through several mechanistic pathways:

  • Metabolic Reprogramming: GM-CSF enhances mitochondrial function in mononuclear phagocytes, supporting increased energy production necessary for their effector functions . This metabolic enhancement is particularly critical during inflammatory responses when energy demands escalate.

  • Augmentation of Phagocytic Capacity: GM-CSF significantly increases the phagocytic activity of macrophages and other myeloid cells, facilitating pathogen clearance and tissue homeostasis through enhanced efferocytosis (clearance of apoptotic cells) .

  • Differentiation Regulation: In the context of placental development, GM-CSF influences trophoblast differentiation by regulating key genes including Ascl2, Tcfeb, Itgav, and Socs3 . This regulatory function extends to myeloid lineage differentiation in other tissues.

  • Cytokine Network Integration: GM-CSF participates in complex "CSF networks" where interdependent coregulation with proinflammatory cytokines like IL-1, TNF, IL-23, and IL-6 creates autocrine/paracrine feedback loops that amplify and sustain inflammatory responses .

  • Cell Survival Promotion: GM-CSF supports the survival of dendritic cells in non-lymphoid tissues, contributing to tissue-resident immune surveillance and homeostasis .

  • Regulation of Cell Trafficking: Evidence suggests GM-CSF may control monocyte-derived population numbers in inflammatory sites by influencing cell trafficking, survival, or even local proliferation .

These mechanisms collectively explain how therapeutic administration of exogenous rhu GM-CSF can potentially correct mononuclear phagocyte dysfunction in diseases characterized by GM-CSF deficiency or insufficiency.

What experimental models are most appropriate for studying GM-CSF function in different disease contexts?

The selection of experimental models for studying GM-CSF function varies depending on the disease context and research questions. Several established models have proven valuable:

  • Csf2 Null Mutation Mouse Models: These knockout models have been instrumental in uncovering GM-CSF's role in placental development and fetal growth. Csf2−/− mice exhibit fetal growth restriction in utero, elevated rates of late gestation fetal loss, and early postnatal mortality . They display altered placental structure with a decreased labyrinthine zone:junctional zone ratio, providing insights into GM-CSF's role in trophoblast differentiation.

  • TF-1 Cell Proliferation Assays: This human erythroleukemia cell line is GM-CSF-dependent and serves as a standard bioassay for quantitatively evaluating GM-CSF biological activity. The proliferative response typically shows an ED50 of 6-30 pg/mL for recombinant human GM-CSF .

  • Primary Human Bone Marrow Cell Cultures: These ex vivo systems allow assessment of GM-CSF effects on multiple hematopoietic lineages simultaneously. Studies have shown that GM-CSF increases both erythroid precursors and granulocytes after 48 hours of incubation .

  • Autoimmune Disease Models: Several inflammatory and autoimmune disease models have been instrumental in evaluating GM-CSF's role and therapeutic potential:

    • Autoimmune arthritis models

    • Experimental autoimmune myocarditis

    • Myocardial infarction models

    • Inflammatory bowel disease models

    • Inflammatory-dilated cardiomyopathy models

  • Inflammatory Challenge Models: Systems such as antigen-induced mouse peritonitis have revealed GM-CSF's role in regulating macrophage and dendritic cell numbers in inflammatory settings .

The choice of model should align with specific research objectives, whether investigating basic GM-CSF biology, therapeutic applications, or disease mechanisms.

What are the current approaches for targeting GM-CSF in inflammatory/autoimmune disease models?

Research into GM-CSF targeting in inflammatory and autoimmune diseases has evolved into several distinct strategic approaches:

  • GM-CSF Neutralization: Antibody-mediated neutralization of GM-CSF has shown efficacy in multiple preclinical models of inflammatory and autoimmune conditions. This approach directly prevents GM-CSF from binding to its receptor, inhibiting downstream signaling .

  • Receptor Blockade: Targeting the GM-CSF receptor (particularly the alpha subunit) represents another approach to interrupt GM-CSF signaling. This strategy may offer advantages when multiple ligands for the receptor exist .

  • GM-CSF Supplementation: Paradoxically, in some models, GM-CSF administration has shown beneficial effects:

    • GM-CSF has improved outcomes in certain inflammatory settings through the promotion of tolerogenic dendritic cells

    • Administration of exogenous GM-CSF to mice has demonstrated protection against embryonic and fetal loss

  • Chimeric/Fusion Proteins: Development of GM-CSF fusion constructs represents an innovative approach to modify GM-CSF functionality. For example, GM-CSF-ApoA-I chimera has shown enhanced efficacy in maintaining bone marrow cell viability and reducing apoptosis compared to authentic GM-CSF .

  • Modification Strategies: Researchers employ various modification strategies to enhance GM-CSF properties:

    • Pegylation: Covalent binding to polyethylene glycol to increase half-life, though this approach can lead to decreased biological activity and potential anti-PEG antibody formation

    • Fusion proteins: Creation of chimeric molecules containing GM-CSF fused to proteins that complement cytokine function and protect from proteolytic degradation

The effectiveness of these approaches varies considerably depending on the disease model, highlighting the context-dependent role of GM-CSF in inflammation.

How should researchers design dose-response experiments with recombinant GM-CSF?

Designing robust dose-response experiments with recombinant GM-CSF requires careful consideration of multiple factors to ensure reproducibility and meaningful results:

  • Selection of Appropriate Recombinant GM-CSF:

    • Consider the expression system (E. coli, yeast, or mammalian) based on research needs

    • Account for glycosylation differences that affect potency

    • Use GMP-grade products when possible for consistent quality

  • Concentration Range Determination:

    • Begin with a wide concentration range spanning at least 5-6 orders of magnitude

    • For TF-1 cell proliferation assays, include the known ED50 range of 6-30 pg/mL

    • For bone marrow cell experiments, higher concentrations may be needed to observe multilineage effects

  • Appropriate Controls:

    • Include negative controls (vehicle only)

    • Use a reference standard calibrated against the WHO International Standard (NIBSC code: 88/646)

    • Consider including a different cytokine control to confirm specificity

  • Readout Selection:

    • Choose readouts appropriate to the biological effect being studied:

      • Proliferation assays (TF-1 cells, bone marrow cells)

      • Flow cytometry for cell differentiation markers

      • Functional assays (phagocytosis, cytokine production)

      • Gene expression analysis

      • Metabolic parameters (mitochondrial function)

  • Time Course Considerations:

    • Include multiple time points to capture both early and late responses

    • For bone marrow cell experiments, 48-hour incubation has shown effects on both erythroid precursors and granulocytes

  • Statistical Design:

    • Perform experiments in triplicate at minimum

    • Calculate EC50/ED50 values using appropriate curve-fitting software

    • Consider reporting both fold change and absolute values

  • Validation Approach:

    • Confirm biological activity through multiple independent assays

    • Verify receptor-dependence using receptor blocking antibodies or inhibitors

Following these methodological considerations will help ensure the generation of reliable and interpretable dose-response data for recombinant GM-CSF.

What methods can be used to assess GM-CSF-dependent cell proliferation and differentiation?

Multiple complementary methods can be employed to comprehensively assess GM-CSF-dependent cell proliferation and differentiation:

  • Cell Proliferation Assays:

    • TF-1 Cell Bioassay: The human erythroleukemia TF-1 cell line is GM-CSF-dependent and serves as a standard for evaluating biological activity. Proliferation can be measured using colorimetric assays (MTT/XTT), direct cell counting, or metabolic indicators .

    • [³H]-Thymidine Incorporation: Measures DNA synthesis as an indicator of proliferation.

    • BrdU Incorporation: An alternative to radioactive thymidine, detectable by flow cytometry or immunohistochemistry.

    • CFSE Dilution: Flow cytometry-based method tracking cell division through sequential dilution of fluorescent dye.

  • Differentiation Assessment:

    • Flow Cytometry: Analysis of lineage markers can identify and quantify different cell populations. For bone marrow cells, erythroid precursors and granulocyte populations can be distinguished and enumerated .

    • Morphological Analysis: Microscopic examination of cell morphology, particularly for assessing neutrophil maturation and segmentation .

    • Colony-Forming Assays: Methylcellulose-based colony assays can identify different progenitor populations (CFU-GM, BFU-E, etc.) responding to GM-CSF.

    • Gene Expression Analysis: qRT-PCR or microarray analysis of lineage-specific genes and differentiation markers. For example, trophoblast differentiation genes like Ascl2, Tcfeb, Itgav, and Socs3 are regulated by GM-CSF in placental development .

  • Functional Readouts:

    • Phagocytosis Assays: Quantify phagocytic capacity using fluorescent particles or labeled bacteria.

    • Cytokine Production: Measure secretion of downstream cytokines via ELISA or multiplex assays.

    • Chemotaxis Assays: Evaluate migratory response to chemotactic stimuli.

    • Cell Viability and Apoptosis: Assess resistance to apoptosis using Annexin V/PI staining or similar methods .

  • Advanced Analytical Approaches:

    • Single-cell RNA Sequencing: Provides high-resolution data on differentiation trajectories and heterogeneity within responding populations.

    • Mass Cytometry (CyTOF): Enables simultaneous analysis of numerous markers to comprehensively characterize differentiation states.

    • Metabolic Profiling: Assesses changes in cellular metabolism associated with GM-CSF stimulation.

These methods provide complementary information and should be selected based on specific research questions and available resources.

What are the optimal conditions for maintaining GM-CSF stability in experimental settings?

Maintaining GM-CSF stability throughout experimental procedures is critical for obtaining reliable results. Research suggests the following optimal conditions:

  • Storage Considerations:

    • Lyophilized Form: Store unopened at -20°C to -80°C for maximum stability .

    • Reconstituted Protein: For short-term use (1-2 weeks), store at 2-8°C; for longer storage, prepare aliquots and store at -20°C to -80°C to avoid repeated freeze-thaw cycles.

    • Working Solutions: Prepare fresh when possible, particularly for long incubation experiments.

  • Reconstitution Parameters:

    • Buffer Selection: Typically, sterile water, PBS, or specialized formulation buffers with a neutral pH (6.8-7.4).

    • Protein Concentration: Higher concentrations often provide better stability; consider using carrier proteins for very dilute solutions.

    • Carrier Proteins: Addition of carrier proteins (0.1-1% BSA or HSA) can prevent adsorption to container surfaces and enhance stability, particularly at low concentrations.

  • Handling Precautions:

    • Temperature Control: Maintain at 2-8°C during experimental setup; avoid prolonged exposure to room temperature.

    • Container Materials: Use low-protein binding materials (polypropylene) for storage and experimental vessels.

    • Gentle Handling: Avoid vigorous agitation or vortexing which can cause protein denaturation; use gentle inversion for mixing.

  • Stability Enhancement Strategies:

    • Protease Inhibitors: Consider adding protease inhibitors when working with complex biological samples.

    • Reducing Agents: For some applications, low concentrations of reducing agents may help maintain protein integrity.

    • Stabilizing Excipients: Compounds like trehalose or glycerol can enhance stability during freeze-thaw cycles.

  • Quality Control Measures:

    • Activity Testing: Periodically verify biological activity using standardized assays like TF-1 cell proliferation.

    • SDS-PAGE Analysis: Monitor for potential degradation using silver staining techniques .

    • MALDI-TOF Analysis: Can be used to confirm molecular mass integrity and detect potential modifications .

Careful attention to these stability parameters will help ensure experimental consistency and reliable results when working with recombinant GM-CSF.

What is the therapeutic potential of recombinant GM-CSF in diseases of GM-CSF insufficiency?

Recombinant human GM-CSF shows significant therapeutic potential across multiple diseases characterized by GM-CSF deficiency or insufficiency:

  • Mononuclear Phagocyte Dysfunction Disorders: Exogenous rhu GM-CSF (e.g., sargramostim) can support and restore the metabolic capacity and function of mononuclear phagocytes, potentially addressing conditions where these cells show functional impairment .

  • Neutropenia and Neutropenic Complications: Recombinant GM-CSF is widely used in the prevention of neutropenia and associated complications, with chimeric forms like GM-CSF-ApoA-I showing enhanced efficacy in normalizing neutrophil proliferation, maturation, and segmentation under conditions of impaired granulopoiesis .

  • Placental Insufficiency: Research in Csf2−/− mice suggests potential applications in addressing placental development issues. Administration of exogenous GM-CSF to mice has demonstrated protection against embryonic and fetal loss, possibly through supporting proper trophoblast differentiation and placental development .

  • Immune Checkpoint Inhibitor Therapy Enhancement: Emerging evidence suggests rhu GM-CSF may augment the anti-cancer effects of immune checkpoint inhibitor immunotherapy while potentially ameliorating immune-related adverse events .

  • Neurodegenerative Disorders: Ongoing research points to potential effects of innate immune system modulation via GM-CSF on patient outcomes in neurodegenerative conditions, though this remains an active area of investigation .

The therapeutic applications of recombinant GM-CSF represent a paradigm shift toward classifying and treating diseases based on cytokine insufficiency, addressing a high unmet medical need across multiple conditions.

How does GM-CSF contribute to inflammatory/autoimmune disease mechanisms?

GM-CSF plays complex and sometimes contradictory roles in inflammatory and autoimmune disease mechanisms:

  • Inflammatory Cell Recruitment and Activation: In inflammatory settings, GM-CSF can drive the recruitment, activation, and survival of myeloid cells, particularly monocytes/macrophages and neutrophils. GM+ neutrophils have been identified as major infiltrating cells in interstitial lung disease in autoimmune arthritis models .

  • Tissue-Specific Sources and Effects: Multiple cell types can produce GM-CSF in inflammatory contexts:

    • Fibroblast-like synoviocytes contribute to autoimmune arthritis initiation

    • Cardiac fibroblast-derived GM-CSF has been implicated in Kawasaki disease and myocarditis

    • Epithelial cells produce GM-CSF in response to allergenic stimuli during allergic sensitization

  • Cytokine Network Integration: GM-CSF participates in the "CSF network," establishing positive feedback loops with other proinflammatory cytokines such as IL-1, TNF, IL-23, and IL-6. These autocrine/paracrine networks involve interactions between macrophages, DCs, and T-helper cells, potentially driving chronic inflammation .

  • Disease-Specific Feedback Mechanisms: Recent research has identified positive feedback loops involving GM-CSF in:

    • Intestinal inflammation

    • Inflammatory-dilated cardiomyopathy

    • Breast cancer metastasis

  • Dual Role in Inflammation: Paradoxically, while GM-CSF exacerbates certain inflammatory models, its administration can also improve outcomes in other contexts through mechanisms such as promoting tolerogenic dendritic cells .

These diverse mechanisms highlight the context-dependent nature of GM-CSF in inflammation and suggest that therapeutic targeting strategies must carefully consider disease-specific pathways.

What are the optimal biomarkers for monitoring GM-CSF activity in experimental and clinical settings?

Effective monitoring of GM-CSF activity requires a multi-parameter approach incorporating both direct and indirect biomarkers:

  • Direct GM-CSF Measurements:

    • Serum/Plasma GM-CSF Levels: Quantified by high-sensitivity ELISA or multiplex assays

    • GM-CSF mRNA Expression: In relevant tissues or circulating cells using qRT-PCR

    • GM-CSF Receptor Expression: Flow cytometric analysis of CSF2RA (CD116) and CSF2RB (CD131) on target cell populations

  • Myeloid Cell Parameters:

    • Neutrophil Counts and Function: Absolute counts, maturation state, and functional assays (oxidative burst, phagocytosis)

    • Monocyte/Macrophage Activation Markers: CD80/86, HLA-DR, CD163, CD206

    • Dendritic Cell Subsets: Enumeration and phenotypic characterization of myeloid DCs

  • Downstream Signaling Indicators:

    • Phosphorylated STAT5: A direct indicator of GM-CSF receptor signaling

    • Metabolic Parameters: Mitochondrial function, glycolytic activity

    • Gene Expression Signatures: Transcriptional profiles of GM-CSF-responsive genes

  • Disease-Specific Response Markers:

    • Placental Development: Monitoring of trophoblast differentiation markers (Ascl2, Tcfeb, Itgav, Socs3) in relevant models

    • Inflammatory Models: Pro-inflammatory cytokine levels (IL-1, TNF, IL-6) that participate in GM-CSF networks

    • Bone Marrow Response: Evaluation of both erythroid precursors and granulocyte populations

  • Functional Readouts:

    • TF-1 Cell Bioassay: For measuring biologically active GM-CSF in research samples

    • In Vivo Mobilization: Assessment of myeloid cell mobilization in response to GM-CSF administration

    • Tissue-Specific Effects: Evaluation of resident myeloid cell populations and their functional state

The selection of appropriate biomarkers should be tailored to the specific research question or clinical application, with consideration of both the direct effects of GM-CSF and its downstream biological consequences.

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.