SCF Canine

Stem Cell Factor Canine Recombinant
Shipped with Ice Packs
In Stock

Description

Stem Cell Factor Canine Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 165 amino acids and having a molecular mass of 18.4kDa Dalton.
The SCF is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Stem cell factor / KIT ligand (SCF), also known as "steel factor" or "c-kit ligand," is a cytokine that binds to CD117 (c-Kit). It exists in two forms: cell surface bound and soluble (or free), the latter resulting from the cleavage of the former by metalloproteases. SCF is crucial for the survival, proliferation, and differentiation of hematopoietic stem cells and other hematopoietic progenitor cells. Notably, it plays a role in the development of erythrocyte precursors by transforming burst-forming unit-erythroid (BFU-E) cells into colony-forming unit-erythroid (CFU-E) cells.
Description
Recombinant Canine Stem Cell Factor, produced in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 18.4 kDa. It comprises 165 amino acids. The purification process involves proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The product is lyophilized from a 0.2 µm filtered solution concentrated in PBS with a pH of 7.4.
Solubility
For reconstitution, it is advised to dissolve the lyophilized Stem Cell Factor in sterile 18 MΩ-cm H2O at a concentration not less than 100 µg/ml. This solution can be further diluted into other aqueous solutions as needed.
Stability
Lyophilized SCF, while stable at room temperature for up to 3 weeks, should ideally be stored desiccated at a temperature below -18°C. Once reconstituted, SCF should be stored at 4°C for a period of 2-7 days. For long-term storage, freezing at -18°C is recommended, and the addition of a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
The purity of the product is determined to be greater than 97.0% using HPLC and SDS-PAGE analysis.
Biological Activity
The biological activity is determined by the dose-dependent stimulation of human TF-1 cell proliferation. The ED50 is less than 2.0 ng/ml, which corresponds to a specific activity of 5,000,000 IU/mg. This indicates full biological activity compared to the standard.
Synonyms
Kit ligand Precursor, C-kit ligand, SCF, Mast cell growth factor, MGF, SF, KL-1, Kitl, DKFZp686F2250.
Source
Escherichia Coli.
Amino Acid Sequence
KGICGKRVTD DVKDVTKLVA NLPKDYKIAL KYVPGMDVLP SHCWISVMVE QLSVSLTDLL DKFSNISEGL SNYSIIDKLV KIVDDLVECT EGYSFENVKK APKSPELRLF TPEEFFRIFN RSIDAFKDLE TVASKSSECV VSSTLSPDKD SRVSVTKPFM LPPVA

Q&A

What is canine SCF and what are its primary biological functions?

Canine Stem Cell Factor (SCF), also known as KIT ligand or mast cell growth factor, is a cytokine that plays crucial roles in canine physiology. SCF stimulates the proliferation of mast cells and augments the proliferation of both myeloid and lymphoid hematopoietic progenitors in bone marrow culture . It also mediates cell-cell adhesion and acts synergistically with other cytokines to regulate various cellular processes .

From a methodological standpoint, researchers studying SCF function typically employ:

  • In vitro colony formation assays to assess effects on hematopoietic progenitors

  • Cell proliferation assays with various concentrations of recombinant canine SCF (rc-SCF)

  • In vivo administration protocols, with effective doses ranging from 10-30 μg/kg/day for long-term treatment

  • Molecular analysis of SCF gene expression in different tissues and disease states

How does the SCF-KIT signaling axis function in canine cells?

SCF binds to its receptor KIT (CD117), which is a membrane-bound receptor tyrosine kinase expressed on various cell types including mast cells, hematopoietic stem cells, interstitial cells of Cajal, melanocytes, and germ cells . This interaction initiates signaling cascades critical for cell survival, proliferation, and differentiation.

Research methodologies to investigate this signaling axis include:

  • Immunohistochemical analysis of KIT expression patterns in tissues

  • Assessment of KIT localization (membranous, cytoplasmic, or perinuclear) which correlates with functional status

  • Analysis of downstream phosphorylation events following SCF binding

  • Characterization of signaling pathway components activated after receptor engagement

The SCF-KIT pathway is particularly significant in canine mast cell biology, where alterations in this signaling pathway are associated with mast cell tumors and their biological behavior .

What experimental systems are used to study SCF effects on canine hematopoiesis?

Several experimental approaches have been established to investigate SCF's role in canine hematopoiesis:

  • Grey collie model: Grey collie dogs have cyclic fluctuations in blood cell counts caused by a regulatory defect of hematopoietic stem cells, making them an excellent natural model for studying hematopoietic regulation .

  • In vitro colony assays: Colony-forming unit granulocyte-macrophage (CFU-GM) formation from canine bone marrow can be assessed in response to rc-SCF, with dose-response curves providing quantitative data on hematopoietic progenitor stimulation .

  • In vivo administration studies: Treating dogs with rc-SCF at various doses (10-100 μg/kg/d) can demonstrate effects on neutrophil production and cycling. At doses of 20 μg/kg/d or greater, rc-SCF abrogates neutropenic periods in grey collies .

  • Combination cytokine studies: The combination of rc-G-CSF (0.5-1.0 μg/kg/d) with rc-SCF (20-50 μg/kg/d) produces synergistic effects on neutrophil levels, demonstrating cooperative interactions between these hematopoietic regulators .

What molecular mechanisms explain SCF's role in canine mast cell tumor development?

The molecular basis of SCF's involvement in canine mast cell tumors (MCTs) centers on its interaction with the KIT receptor and associated signaling pathways:

  • c-kit mutations: Mutations in the c-kit gene, which encodes the KIT receptor, have been identified in canine MCTs and affect prognosis . These mutations can lead to constitutive (ligand-independent) receptor activation.

  • KIT expression patterns: Immunohistochemical analysis of KIT localization patterns in MCTs provides prognostic information. Aberrant cytoplasmic or perinuclear KIT localization often correlates with more aggressive tumor behavior compared to normal membranous expression .

  • Breed predispositions: Certain breeds show higher incidence of MCTs, including Boxers, Golden and Labrador Retrievers, French Bulldogs, and American Staffordshire Terriers . Interestingly, French Bulldogs—typically developing benign variants at other skin sites—were more likely to have poorly differentiated digital MCTs in one study .

Research methodologies for investigating these mechanisms include immunohistochemistry for KIT expression, PCR-based mutation detection in c-kit, and correlation of molecular findings with histological grade and clinical outcomes.

How can SCF levels be accurately quantified in canine clinical samples?

Several validated methodologies exist for measuring SCF in canine samples:

Protein-level detection methods:

  • ELISA: Commercial sandwich (quantitative) ELISA kits specifically developed for canine SCF measurement show reliable performance across sample types :

Sample typeRecovery (Average %)Range
Serum104.8%86-123%
Plasma101.2%84-115%
Cell culture media96.16%87-104%
  • Immunohistochemistry: For tissue localization of SCF protein, with macrophages identified as a source of SCF in infarcted myocardium .

mRNA-level detection methods:

  • RT-PCR: Used to obtain canine-specific SCF clones encoding nucleotides 1-795 of the published canine SCF sequence .

  • Nuclease protection assay: Demonstrated induction of SCF mRNA within 72 hours of reperfusion in a canine myocardial infarction model .

For optimal results, sample collection and handling protocols must be standardized, with consideration of potential confounding factors such as platelet activation that might release SCF during processing.

What is the significance of SCF in canine cardiovascular disease models?

SCF plays important roles in myocardial healing following ischemic injury, as demonstrated in canine models:

  • Mast cell recruitment: Following myocardial infarction, there is a striking increase in mast cell numbers during the healing phase. Mast cell numbers begin increasing after 72 hours of reperfusion, with maximum accumulation in areas of collagen deposition (12.0±2.6-fold increase; P<0.01) .

  • SCF expression by macrophages: Immunohistochemical studies have demonstrated that a subset of macrophages is the source of SCF immunoreactivity in infarcted myocardium. SCF protein was not found in endothelial cells or myofibroblasts .

  • Mast cell precursor infiltration: Intravascular tryptase-positive, FITC-avidin-positive, CD11b-negative mast cell precursors were observed in healing areas and cardiac lymph after 48-72 hours of reperfusion .

  • SCF induction timing: Using a nuclease protection assay, SCF mRNA was shown to be induced within 72 hours of reperfusion .

These findings suggest that SCF produced by macrophages promotes chemotaxis and growth of mast cell precursors in the healing heart, indicating an important role in cardiac repair processes.

How do SCF-based therapeutic approaches compare with other cytokine treatments in canine models?

SCF-based therapies offer unique advantages and synergistic potential when compared with other cytokine treatments:

  • Monotherapy effectiveness: Daily rc-SCF administration at doses ≥20 μg/kg/d successfully abrogates neutropenic periods in grey collies with cyclic hematopoiesis .

  • Dose-response relationship: While lower doses (10-30 μg/kg/d) are generally well-tolerated for long-term treatment, higher doses (100 μg/kg/d) can induce neutrophilia, indicating a dose-dependent effect on neutrophil production .

  • Synergistic combinations: When rc-SCF (20-50 μg/kg/d) is combined with rc-G-CSF (0.5-1.0 μg/kg/d), the resulting neutrophil levels are higher than the sum of levels when these cytokines are given separately . This synergistic effect suggests distinct but complementary mechanisms of action.

  • Long-term tolerability: Unlike some cytokine therapies that may induce neutralizing antibodies or tachyphylaxis, long-term treatment with rc-SCF at appropriate doses is generally well-tolerated in canine models .

This evidence suggests that SCF may be particularly valuable in therapeutic strategies for chronic hypoproliferative disorders of hematopoiesis, either alone or as part of combination approaches.

What genetic factors influence SCF expression and function in different canine breeds?

While research on breed-specific variations in SCF biology is still developing, several genetic considerations are relevant:

  • SCF gene variants: Analysis of the SCF gene in grey collies with cyclic hematopoiesis showed no evidence of mutations in the coding region , suggesting that the primary defect in this condition lies elsewhere in the hematopoietic regulatory network.

  • Breed predispositions to SCF-responsive tumors: The frequency of mast cell tumors varies significantly across breeds, with Boxers, Golden and Labrador Retrievers, French Bulldogs, and American Staffordshire Terriers being particularly susceptible . These breed predispositions suggest genetic factors that may interact with SCF-KIT signaling.

  • c-kit mutation patterns: Different patterns of c-kit mutations may occur with varying frequencies across breeds. These mutations can affect KIT's response to SCF, altering downstream signaling pathways .

  • Breed-specific tumor behavior: French Bulldogs typically develop benign mast cell tumors at most skin sites, but interestingly were found to have a higher likelihood of poorly differentiated tumors when MCTs occurred on the digits . This suggests complex breed-specific interactions between genetic factors and SCF-KIT signaling.

Research methodologies to investigate these factors include comparative genomic analyses across breeds, screening for polymorphisms in SCF and KIT genes, and correlating genetic findings with clinical observations.

What are the optimal protocols for producing and validating recombinant canine SCF?

The production and validation of high-quality recombinant canine SCF (rc-SCF) for research applications involves several critical steps:

  • Expression systems: Bacterial, insect, or mammalian cell expression systems can be used, with each offering different advantages for post-translational modifications. The specific expression system should be selected based on the intended application.

  • Purification strategies: Multi-step purification protocols typically include affinity chromatography, size exclusion, and endotoxin removal steps to ensure high purity.

  • Biological activity testing: Validation through dose-response curves in canine bone marrow colony formation assays . CFU-GM formation from normal and affected dogs should show similar dose-response patterns to confirm biological activity.

  • Quality control parameters:

    • Purity assessment by SDS-PAGE and HPLC

    • Endotoxin testing to ensure levels are below thresholds that could confound experimental results

    • Stability testing under various storage conditions

  • In vivo validation: Confirmation of biological effects in canine models, such as demonstrating that rc-SCF abrogates neutropenic periods in grey collies at appropriate doses .

These methodological considerations are essential for ensuring consistent and reliable results in both basic research and potential therapeutic applications.

What experimental design factors are critical when studying SCF in canine disease models?

When designing experiments to investigate SCF in canine disease models, researchers should consider several key factors:

  • Control selection: Appropriate controls are essential, including age-matched, breed-matched, and sex-matched controls when possible. In studies of grey collie dogs with cyclic hematopoiesis, comparing to normal dogs provides crucial insights .

  • Timing of measurements: For conditions with cyclical patterns (like cyclic hematopoiesis) or dynamic disease processes (like myocardial infarction), the timing of sample collection and analysis is critical. For example, SCF mRNA induction occurs within 72 hours of reperfusion in myocardial infarction models .

  • Dose optimization: When administering rc-SCF, dose-response relationships must be established. Studies have shown that doses of 20 μg/kg/d or greater are needed to abrogate neutropenic periods in grey collies .

  • Sample size calculations: Statistical power analysis should guide determination of appropriate sample sizes, considering the variability in the parameters being measured.

  • Multi-parameter assessment: Comprehensive evaluation should include measures of SCF protein and mRNA levels, receptor expression and localization, downstream signaling events, and functional outcomes.

  • Longitudinal design: For chronic conditions or treatment studies, longitudinal assessment allows tracking of changes over time and evaluation of long-term effects and tolerability .

Addressing these design factors enhances the rigor and reproducibility of research on SCF in canine disease models.

How can researchers effectively compare canine and human SCF biology for translational studies?

Comparative studies of canine and human SCF biology require specific methodological approaches to generate translational insights:

  • Sequence and structural comparisons: Alignment of canine and human SCF amino acid sequences to identify conserved and divergent regions. Structural modeling based on sequence data can predict functional similarities and differences.

  • Cross-species activity testing: Determining whether human SCF can activate canine KIT receptor and vice versa, and comparing relative potencies.

  • Signaling pathway conservation analysis: Comparative assessment of downstream signaling events triggered by SCF in canine versus human cells, identifying shared and species-specific pathways.

  • Disease model parallels: Evaluating whether SCF plays similar roles in analogous diseases across species, such as comparing canine and human mast cell disorders.

  • Pharmacokinetic and pharmacodynamic comparisons: Analyzing differences in metabolism, distribution, and biological effects of SCF between species to inform dosing for therapeutic applications.

  • One medicine approach: As highlighted in regenerative medicine research, the development of therapies for human patients can lead to innovative treatments for animals, while pre-clinical studies in animals provide knowledge to advance human medicine .

These comparative approaches facilitate bidirectional translation between canine and human research, ultimately benefiting both veterinary and human medicine.

What are the potential applications of SCF in regenerative medicine for canines?

SCF shows promise for several applications in canine regenerative medicine:

  • Hematopoietic recovery: Based on its ability to abrogate neutropenic periods in grey collies , SCF could potentially accelerate hematopoietic recovery following myelosuppressive therapies like chemotherapy or radiation.

  • Tissue repair enhancement: Given SCF's role in mast cell recruitment to areas of myocardial injury , it may have applications in enhancing tissue repair processes in various organs.

  • Stem cell mobilization: SCF could be used to mobilize stem cells for collection and use in regenerative therapies, similar to applications being explored in human medicine.

  • Combined approaches with iPSCs: While not directly mentioned in the search results, SCF might have applications in conjunction with induced pluripotent stem cells (iPSCs), which are being developed for veterinary species . SCF could potentially enhance the differentiation or engraftment of iPSC-derived cells.

  • Synergistic combinations: The demonstrated synergy between SCF and G-CSF suggests that combination approaches may be particularly effective in regenerative applications.

These potential applications warrant further investigation through well-designed preclinical studies and eventually clinical trials in canine patients.

What technological advances could improve the study of SCF signaling in canine tissues?

Several emerging technologies hold promise for advancing our understanding of SCF signaling in canine tissues:

  • Single-cell analysis techniques: Single-cell RNA sequencing and mass cytometry could provide unprecedented insights into heterogeneity of SCF production and response within tissues.

  • Spatial transcriptomics and proteomics: These approaches could map the distribution of SCF, KIT, and associated signaling molecules within complex tissue environments with high spatial resolution.

  • CRISPR-based screening: Genome-wide CRISPR screens in canine cell lines could identify novel regulators and effectors in the SCF-KIT signaling pathway.

  • Phosphoproteomics: Comprehensive analysis of phosphorylation events downstream of KIT activation could elucidate the full complexity of SCF signaling networks.

  • In vivo imaging techniques: Development of traceable SCF molecules or reporter systems could allow real-time visualization of SCF distribution and activity in living animals.

  • Canine-specific antibodies and detection reagents: Improved availability of canine-specific research tools would enhance the sensitivity and specificity of SCF-related studies.

These technological advances would facilitate more comprehensive and precise analysis of SCF biology in normal and diseased canine tissues.

Product Science Overview

Biological Significance

SCF is essential for the survival, proliferation, and differentiation of hematopoietic stem cells, which are the precursors to all blood cell types. It also supports the migration and proliferation of mesenchymal stem cells (MSCs), which can differentiate into various cell types such as bone, cartilage, and fat cells . This makes SCF a vital component in regenerative medicine and cellular therapies.

Production and Purification

The recombinant canine SCF is produced in E. coli and purified using proprietary chromatographic techniques to ensure high purity and activity . The protein is typically lyophilized from a filtered solution in phosphate-buffered saline (PBS) and can be reconstituted in sterile PBS for use in various applications .

Applications in Research and Medicine

Recombinant canine SCF is widely used in research to study stem cell biology, hematopoiesis, and tissue regeneration. It is also employed in preclinical and clinical settings to enhance the efficacy of stem cell therapies. For instance, SCF has been shown to support the migration and proliferation of canine mesenchymal stem cells, making it a valuable tool in veterinary regenerative medicine .

Storage and Stability

The stability and storage conditions for recombinant canine SCF are critical to maintaining its activity. The lyophilized protein should be stored at -20 to -70°C and reconstituted in sterile conditions to prevent contamination. Once reconstituted, the protein can be stored at 2 to 8°C for up to one month or at -20 to -70°C for up to three months under sterile conditions .

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.