GM CSF K9

Granulocyte Macrophage-Colony Stimulating Factor Canine Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

GM-CSF K9 regulates:

  • Myeloid cell proliferation: Stimulates granulocyte, macrophage, and eosinophil production

  • Immune activation: Enhances antigen presentation via dendritic cell maturation

  • Cellular communication: Coordinates hematopoietic niche signaling through paracrine/autocrine mechanisms

Notably, human GM-CSF demonstrates cross-species activity in canine cells despite sequence variations .

Therapeutic Applications

Recent studies demonstrate clinical potential in:

A. Oncolytic Virotherapy
Persistent CDV-Ondneon-GM-CSF infection in DH82 cells:

ParameterResult
GM-CSF secretion1,200-1,800 pg/mL (stable at pH 2-9)
Tumor infiltration↑ CD4+/CD8+ T cells (p<0.01)
Viral persistence>30 passages without attenuation

This engineered virus showed significant anti-tumor effects in canine histiocytic sarcoma models .

B. Vaccine Adjuvants
In DNA vaccine trials:

  • 4.8-fold ↑ IFN-γ secretion vs controls (p<0.001)

  • Neutralizing antibody titers sustained for 56 days post-immunization

  • Species specificity: While human GM-CSF activates canine receptors, reverse compatibility remains untested

  • Dual-phase activity: Pro-tumoral effects observed at high concentrations (>50 ng/mL)

  • Delivery optimization: Sustained release formulations show 38% improved bioavailability vs bolus injections

Emerging solutions include nanoparticle encapsulation and viral vector delivery systems showing promise in preclinical trials .

Product Specs

Introduction
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that plays a crucial role in the production, differentiation, and function of granulocytes and macrophages, which are essential components of the immune system. The active form of GM-CSF exists as a homodimer in the extracellular space. The gene encoding GM-CSF is located within a cluster of related genes on chromosome 5q31. This region is known to be frequently involved in interstitial deletions associated with 5q- syndrome and acute myelogenous leukemia. Notably, this gene cluster also harbors genes responsible for encoding other interleukins, including interleukins 4, 5, and 13. GM-CSF exerts its biological effects by stimulating the growth and differentiation of hematopoietic precursor cells originating from various lineages. These lineages encompass granulocytes, macrophages, eosinophils, and erythrocytes, highlighting its broad impact on hematopoiesis.
Description
Recombinant Canine Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF), produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 128 amino acids. With a molecular weight of 14.2 kDa, this protein exhibits high purity, achieved through proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile.
Formulation
The protein was lyophilized following extensive dialysis in 1xPBS at a pH of 7.4.
Solubility
To reconstitute the lyophilized GM-CSF, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions as required.
Stability
Lyophilized GM-CSF demonstrates stability at room temperature for a period of 3 weeks. However, for long-term storage, it is recommended to store the lyophilized product in a desiccated state at a temperature below -18°C. Once reconstituted, GM-CSF should be stored at 4°C for a period of 2-7 days. For extended storage, it is advisable to add a carrier protein, such as 0.1% HSA or BSA, and store the solution below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity of this product is greater than 96.0%, as determined by two methods: 1. Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis. 2. Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The ED50, determined by measuring the dose-dependent stimulation of human TF1 erythroleukemic cell proliferation, typically falls within the range of 1-4 ng/ml.
Synonyms

CSF-2, MGI-1GM, GM-CSF, Pluripoietin-alpha, MGC131935, MGC138897.

Source
Escherichia Coli.
Amino Acid Sequence
APTRSPTLVT RPSQHVDAIQ EALSLLNNSN DVTAVMNKAV KVVSEVFDPE
GPTCLETRLQ LYKEGLQGSL TSLKNPLTMM ANHYKQHCPP TPESPCATQN
INFKSFKENL KDFLFNIPFD CWKPVKK.

Q&A

What is canine GM-CSF and how does it compare structurally to human GM-CSF?

Canine granulocyte-macrophage colony-stimulating factor (caGM-CSF) is a glycoprotein that plays a crucial role in regulating immune cell development and function in dogs. The canine GM-CSF cDNA is 850 base pairs (bp) long and encodes a peptide of 144 amino acids . Comparative analysis shows that the nucleotide and amino acid sequence homology between canine GM-CSF and human GM-CSF (hGM-CSF) is 80% and 70%, respectively . This significant homology indicates conservation of function across species while maintaining species-specific differences that necessitate species-specific reagents for optimal research outcomes.

What are the primary signaling pathways associated with GM-CSF activity?

GM-CSF signaling occurs through a 2-subunit receptor comprising a ligand-specific alpha chain and a common beta chain that is shared with IL-3 and IL-5 . The downstream signaling of GM-CSF receptor (GM-CSFR) is mediated through multiple pathways including:

  • Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5 (STAT5)

  • Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)

  • Extracellular signal-regulated kinase (ERK)

  • Phosphoinositide 3-kinase (PI3K)-Akt pathway

These signaling pathways collectively regulate cell survival, proliferation, differentiation, and functional activation of target cells.

What expression systems are most effective for producing recombinant canine GM-CSF?

Based on the research literature, two main expression systems have been successfully used for canine GM-CSF:

  • Mammalian expression systems: The mammalian expression vector pCMV/CAGM has been successfully used to transfect COS cells for expression of canine GM-CSF . This system produces biologically active caGM-CSF that demonstrates significant stimulating activity in granulocyte-macrophage colony forming unit (CFU-GM) assays of canine marrow.

  • Bacterial expression systems: Bacterial expression of caGM-CSF has also been reported, with the resulting product being functionally active when administered to dogs in vivo .

Additionally, viral vector systems have been employed, where canine distemper virus has been genetically engineered to express canine GM-CSF (CDV-Ond-neon-GM-CSF) .

What methodologies are most reliable for detecting and quantifying GM-CSF in canine samples?

Several methodologies have been validated for detecting and quantifying canine GM-CSF:

  • Immunoblotting: Western blot analysis using primary antibodies directed against canine GM-CSF (e.g., goat polyclonal antibodies at 1:1000 dilution; R&D Systems) with HRP-conjugated secondary antibodies . Protein bands can be visualized using chemiluminescent substrates and quantified through densitometric analysis.

  • Functional bioassays: Activity of canine GM-CSF can be quantified using cell proliferation assays with GM-CSF-responsive cell lines. For example, HeLa cells have demonstrated increased proliferation in response to canine GM-CSF in cell duplication assays .

  • Receptor binding assays: Detection of GM-CSF receptor (CD116) expression using specific antibodies (e.g., rabbit polyclonal antibodies) can help assess potential cellular responsiveness to GM-CSF .

What are the observed hematologic effects of canine GM-CSF administration in dogs?

Administration of canine GM-CSF in dogs produces several significant hematologic effects:

  • Leukocyte populations: Subcutaneous administration of bacterially-expressed caGM-CSF twice daily for 14-16 days results in significant increases in circulating blood neutrophils and monocytes. The effect on eosinophils is more variable .

  • Thrombocytopenia: A notable side effect is the development of thrombocytopenia during GM-CSF administration, which resolves rapidly after treatment cessation. Evaluation of survival times of 51Cr-labeled autologous platelets suggests that increased consumption is the primary mechanism for this thrombocytopenia rather than decreased production .

  • Dose-response relationship: The hematologic effects of GM-CSF administration demonstrate dose-dependency, suggesting careful titration is needed when designing experiments .

These findings highlight the importance of careful monitoring of complete blood counts in canine studies involving GM-CSF administration.

How does GM-CSF influence cancer progression and treatment outcomes in canine models?

GM-CSF has demonstrated complex effects in cancer research that can be both beneficial and potentially detrimental:

Anti-tumoral effects:

  • Direct inhibitory effects on certain tumor cells by inducing cell cycle arrest at the G0/G1 phase

  • Modulation of immune responses against tumor cells

  • Inhibition of angiogenesis

  • Enhancement of both humoral and cellular immunity against tumors

Pro-tumoral concerns:

  • Some studies have reported that GM-CSF may accelerate tumor cell proliferation in certain contexts

  • Enhanced neutrophil recruitment may contribute to lung damage and acute respiratory distress syndrome (ARDS) in some settings

These contrasting effects highlight why GM-CSF is sometimes described as a "double-edged sword" in cancer therapy, necessitating careful experimental design and monitoring.

What methodologies are most appropriate for studying GM-CSF effects in canine histiocytic sarcoma models?

Research with canine histiocytic sarcoma (HS) models, particularly using DH82 cells, has employed several methodological approaches:

  • Cell duplication assays: To assess the effect of GM-CSF on cell proliferation. Commercially available canine GM-CSF is typically supplemented at 5 μg/mL for these experiments .

  • Scratch assays: To evaluate the effect of GM-CSF on cell motility and migration capabilities .

  • Viral vector delivery systems: Using genetically engineered canine distemper virus (CDV) strains to express GM-CSF directly in HS cells .

  • pH stability testing: Acidification of supernatants (pH 2.0 for 30 minutes) to inactivate virus particles while preserving GM-CSF activity for functional studies .

Importantly, research has shown that while GM-CSF produced by CDV-Ond-neon-GM-CSF-infected DH82 cells significantly increases proliferation of HeLa cells, it does not increase proliferation or motility of DH82 cells themselves , suggesting cell-type specific responses.

What controls should be included when evaluating GM-CSF effects in canine studies?

Proper experimental controls are essential for rigorous evaluation of GM-CSF effects:

  • Negative controls:

    • Medium-only conditions

    • Supernatants from non-infected cells

    • Viral vectors lacking the GM-CSF gene (e.g., CDV-Ond and CDV-Ond-neon when studying CDV-Ond-neon-GM-CSF)

  • Positive controls:

    • Commercially available recombinant canine GM-CSF (caGM-CSF)

    • Recombinant human GM-CSF (rhGM-CSF) for comparative studies

  • Treatment validation controls:

    • Confirmation of GM-CSF production through immunoblotting

    • Verification of viral infection where applicable (e.g., detection of CDV nucleoprotein)

    • Receptor expression analysis to confirm cellular capability to respond to GM-CSF

  • Time-course controls:

    • Multiple time points (e.g., 6h, 12h, 24h) to capture both early and late effects

How can GM-CSF stability be ensured during experimental manipulations?

GM-CSF stability is a critical consideration for experimental success:

  • pH stability: Research has demonstrated that canine GM-CSF maintains its biological activity after acidification (pH 2.0 for 30 minutes), with no significant differences in GM-CSF concentration between native and acidified samples . This pH stability is advantageous for experiments requiring virus inactivation while preserving cytokine function.

  • Storage conditions: While specific storage data for canine GM-CSF is limited in the provided sources, typical cytokine storage recommendations include:

    • Aliquoting to avoid freeze-thaw cycles

    • Storage at -80°C for long-term preservation

    • Addition of carrier proteins (e.g., BSA) to prevent adherence to tubes and loss of protein

  • Activity confirmation: Functional validation using bioassays (e.g., HeLa cell proliferation) rather than relying solely on protein concentration measurements .

How can canine GM-CSF be effectively incorporated into viral vector systems for cancer therapy?

The incorporation of canine GM-CSF into viral vectors for cancer therapy involves several strategic considerations:

  • Vector selection: Canine distemper virus (CDV) has been successfully engineered to express canine GM-CSF, creating strains such as CDV-Ond-neon-GM-CSF . This approach parallels the successful use of GM-CSF in other oncolytic viruses, including:

    • Herpes simplex virus (Talimogene laherparepvec/T-VEC, FDA-approved)

    • Adenovirus

    • Vaccinia virus

    • Measles virus

  • Expression confirmation: Verification of GM-CSF expression through:

    • Immunoblotting using antibodies specific to canine GM-CSF

    • Functional assays demonstrating biological activity of the produced GM-CSF

  • Safety considerations: Acidification protocols (pH 2.0) can inactivate viral particles while preserving GM-CSF activity, ensuring that observed effects are attributable to GM-CSF rather than viral activity .

  • Target cell specificity: Assessment of GM-CSF effects on both target cancer cells and non-target cells to evaluate potential off-target effects .

What are the mechanisms by which GM-CSF enhances anti-tumor immunity in canine models?

GM-CSF enhances anti-tumor immunity through multiple mechanisms:

  • Immune cell maturation and differentiation: GM-CSF promotes the maturation and differentiation of:

    • Granulocytes (neutrophils and eosinophils)

    • Monocytes

    • Antigen-presenting cells

    • Indirect effects on CD4+ and CD8+ T lymphocytes

  • Macrophage polarization: GM-CSF influences the polarization of tumor-associated macrophages (TAMs) toward pro-inflammatory phenotypes .

  • Enhanced antigen presentation: By stimulating dendritic cell function, GM-CSF improves tumor antigen presentation and subsequent T-cell activation.

  • Tumor microenvironment modulation: GM-CSF alters the tumor microenvironment to favor anti-tumor immune responses.

  • Long-term immunity development: Studies in murine models have demonstrated that GM-CSF treatment can provide long-term protection against tumor re-engraftment, suggesting the development of immunological memory .

How should researchers address variability in GM-CSF responses between different canine cell types?

The research literature indicates significant variability in GM-CSF responses across different cell types:

  • Receptor expression analysis: Quantify GM-CSF receptor (CD116) expression levels on target cells using techniques such as immunoblotting or flow cytometry to predict responsiveness .

  • Dose-response experiments: Conduct systematic dose-response studies for each cell type to determine optimal GM-CSF concentrations.

  • Time-course evaluations: Assess responses at multiple time points, as some effects may be delayed or transient. For example, HeLa cells showed significant proliferation differences after 12 hours but not after 6 hours of GM-CSF exposure .

  • Cell-specific readouts: Select appropriate readouts for each cell type:

    • Proliferation assays for cells expected to undergo mitosis

    • Migration assays (scratch assays) for cells expected to show motility changes

    • Differentiation markers for cells expected to undergo maturation

  • Mixed culture systems: Consider co-culture systems to better recapitulate in vivo complexity when evaluating immune-mediated effects.

What are the limitations of current canine GM-CSF research models?

Current research models for studying canine GM-CSF have several limitations:

Researchers should consider these limitations when designing experiments and interpreting results from canine GM-CSF studies.

What emerging applications of canine GM-CSF warrant further investigation?

Several promising research directions for canine GM-CSF deserve further exploration:

  • Oncolytic virus combinations: Further development of GM-CSF-expressing canine distemper virus for histiocytic sarcoma treatment , potentially in combination with other immunomodulatory approaches.

  • Comparative oncology models: Using canine GM-CSF studies to inform human cancer therapies, leveraging the advantages of spontaneous canine cancers as models for human disease.

  • Optimized delivery systems: Development of targeted delivery methods to maximize local GM-CSF effects while minimizing systemic side effects such as thrombocytopenia .

  • Biomarker development: Identification of predictive biomarkers for GM-CSF response to better select patients/cases likely to benefit from GM-CSF-based therapies.

  • Combination therapies: Investigation of synergistic effects between GM-CSF and other immunotherapeutic approaches, including checkpoint inhibitors, cancer vaccines, or adoptive cell therapies.

How might techniques from human GM-CSF research be adapted for canine studies?

Adaptation of human GM-CSF research techniques to canine studies could include:

  • Single-cell analysis: Application of single-cell RNA sequencing to better understand the heterogeneity of GM-CSF responses across different immune and cancer cell populations.

  • In vivo imaging: Adaptation of methods to track GM-CSF-responsive cells in living animals to better understand trafficking and localization.

  • Receptor signaling analysis: More detailed examination of species-specific differences in GM-CSF receptor signaling cascades to optimize therapeutic approaches.

  • Combinatorial screening: Systematic evaluation of GM-CSF in combination with other cytokines and therapeutic agents to identify optimal combinations.

  • Translational biomarkers: Development of parallel biomarkers in canine and human patients to facilitate cross-species translation of research findings.

These adaptations would leverage the strengths of canine models while incorporating cutting-edge techniques from human research, potentially accelerating progress in both fields.

Product Science Overview

Discovery and Cloning

The gene for canine GM-CSF has been successfully cloned, and recombinant protein is available for experimental purposes . This recombinant form, known as Granulocyte Macrophage-Colony Stimulating Factor Canine Recombinant, has been instrumental in various research and therapeutic applications.

Function and Mechanism

GM-CSF promotes the survival and activation of macrophages, neutrophils, and eosinophils. It also aids in the maturation of dendritic cells . In vitro studies have shown that canine GM-CSF stimulates the production of granulocyte colony-forming units . When administered in vivo, it causes leukocytosis characterized by mature neutrophilia and, in some cases, monocytosis .

Therapeutic Applications

Canine GM-CSF has shown effectiveness in alleviating severe neutropenia in dogs with cyclic hematopoiesis and post-chemotherapy . The recombinant form has been used experimentally to treat various conditions, demonstrating its potential in veterinary medicine.

Research and Development

Recent studies have explored the role of GM-CSF in autoimmune diseases, revealing its pro-inflammatory functions and contribution to the pathogenicity of Th17 cells . This has led to the development of several agents targeting GM-CSF for the treatment of autoimmune diseases, with promising results in clinical trials .

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.