GMCSF Poricne, His

Granulocyte Macrophage-Colony Stimulating Factor Porcine Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

GMCSF Poricne Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 148 amino acids (18-144a.a.) and having a molecular mass of 16.6kDa.
GMCSF is fused to a 21 amino acid His tag at N-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that stimulates the development of neutrophils and macrophages. It also enhances the proliferation and development of early erythroid, megakaryocytic, and eosinophilic progenitor cells. GM-CSF is secreted by fibroblasts, monocytes, T-lymphocytes, and endothelial cells. This protein blocks the migration of neutrophils and induces the biological activity of mature end-cells.
Description
Recombinant GMCSF Poricne, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 148 amino acids (18-144a.a.). It has a molecular mass of 16.6 kDa. The protein is fused to a 21 amino acid His tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile filtered, colorless solution.
Formulation
GMCSF protein solution (0.5 mg/ml) in Phosphate-Buffered Saline (pH 7.4).
Stability
Store at 4°C if the entire vial will be used within 2-4 weeks. For longer periods, store frozen at -20°C. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid multiple freeze-thaw cycles.
Purity
Greater than 95.0% as determined by SDS-PAGE.
Biological Activity
The ED50 is approximately 40 ng/ml, measured in a cell proliferation assay using TF-1 human erythroleukemic cells.
Synonyms

CSF-2, MGI-1GM, GM-CSF, Pluripoietin-alpha, Molgramostin, Sargramostim

Source

Escherichia Coli.

Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MAPTRPPSPV TRPWQHVDAI KEALSLLNNS NDTAAVMNET VDVVCEMFDP QEPTCVQTRL NLYKQGLRGS LTRLKSPLTL LAKHYEQHCP LTEETSCETQ SITFKSFKDS LNKFLFTIPF DCWGPVKK

Q&A

What is porcine GM-CSF and what are its primary immunological functions?

Porcine Granulocyte-Macrophage Colony-Stimulating Factor (pGM-CSF) is a cytokine first identified as an inducer of differentiation and proliferation of granulocytes and macrophages derived from hematopoietic progenitor cells. It participates in diverse biological processes associated with both innate and adaptive immunity. pGM-CSF is produced by various cell types including immune cells, fibroblasts, and endothelial cells, making it an important regulator of immune responses in pigs. The primary functions of pGM-CSF include regulating proliferation and differentiation of myeloid lineage cells, modulating inflammatory responses, and influencing macrophage activation states, which collectively affect host responses to pathogens like Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) .

How does pGM-CSF expression differ between cell types and under various stimulation conditions?

Expression patterns of pGM-CSF vary significantly between different porcine immune cells and under different stimulation conditions. Studies using real-time quantitative PCR have shown that in porcine alveolar macrophages (PAMs), pGM-CSF expression peaks at approximately 5 hours after lipopolysaccharide (LPS) stimulation and then continuously decreases in the late phase. In contrast, porcine spleen cells show different kinetics depending on the stimulus: LPS-stimulated cells exhibit peak expression at 5 hours, while phytohemagglutinin (PHA)- and concanavalin A (ConA)-stimulated cells show a more gradual increase with peak expression at 24 hours. This cell type-specific and stimulus-dependent expression pattern highlights the complexity of pGM-CSF regulation in the porcine immune system .

What are the current methods for detecting pGM-CSF in biological samples?

Several methods have been developed for detecting pGM-CSF in biological samples, with varying levels of sensitivity and specificity. At the protein level, double-antibody sandwich ELISA incorporating mouse monoclonal antibody (mAb) and rabbit polyclonal antibody against pGM-CSF provides a highly sensitive method for quantifying pGM-CSF in cell culture supernatants and serum samples. For serum analysis, samples typically require a 20-fold dilution in PBS-T buffer before ELISA testing. At the mRNA level, real-time quantitative PCR following RNA isolation and reverse transcription offers a reliable approach for measuring pGM-CSF gene expression. This method is particularly useful for studying expression kinetics in different cell types and under various stimulation conditions. Western blotting can also be employed, though it may have lower sensitivity compared to ELISA for detecting secreted pGM-CSF in biological fluids .

How is His-tagged pGM-CSF expressed and purified for research applications?

Expression and purification of His-tagged pGM-CSF for research applications typically follows a bacterial expression system approach. The process begins with amplification of the pGM-CSF sequence from porcine peripheral blood mononuclear cells (PBMCs) stimulated with LPS. The amplified sequence is then ligated into an expression vector such as pET-28a, which encodes a fusion protein with an 8×His-tag at the C-terminus. This construct is transformed into Escherichia coli strain BL21 (DE3) for recombinant protein expression. Following bacterial culture and induction, the recombinant His-tagged pGM-CSF is purified using affinity chromatography with nickel columns that bind the histidine tag. This method allows for efficient purification of the target protein from bacterial lysates. The resulting purified His-tagged pGM-CSF can then be used for various research applications, including functional assays, structural studies, and as standards for quantitative analyses .

How does post-transcriptional regulation affect pGM-CSF expression during viral infection?

Post-transcriptional regulation significantly impacts pGM-CSF expression during viral infection, particularly PRRSV infection. Research has revealed a striking discrepancy between pGM-CSF mRNA and protein levels in PRRSV-infected porcine alveolar macrophages (PAMs). While PRRSV infection induces higher levels of pGM-CSF mRNA expression compared to non-infected cells, the corresponding protein is often undetectable by Western blot or ELISA. This suggests that PRRSV infection mechanistically exerts post-transcriptional inhibition of pGM-CSF production, effectively blocking the translation of pGM-CSF mRNA into protein. This phenomenon appears to be part of the virus's immune evasion strategy, potentially preventing the antiviral effects that pGM-CSF might otherwise exert. The exact mechanisms of this post-transcriptional control remain to be fully elucidated but could involve viral interference with host translation machinery, enhanced degradation of specific mRNAs, or disruption of protein processing and secretion pathways .

How can transcriptome analysis enhance our understanding of pGM-CSF signaling in PAMs?

Transcriptome analysis offers powerful insights into pGM-CSF signaling in porcine alveolar macrophages (PAMs). Research has shown that increased serum pGM-CSF levels correlate with activation of downstream pGM-CSF signaling in PAMs, as evidenced by an M1-like (pro-inflammatory) gene expression pattern. This approach allows researchers to identify signature genes and pathways activated by pGM-CSF, providing a more comprehensive understanding of its biological effects than single-gene analyses. Transcriptome analysis can reveal how pGM-CSF influences macrophage polarization, antiviral responses, and inflammatory mediator production in the context of viral infections like PRRSV. Furthermore, comparing transcriptomes from animals with different pGM-CSF levels or from cells treated with recombinant pGM-CSF can help identify key regulatory networks and potential therapeutic targets. This approach has particular value when protein-level detection of pGM-CSF is challenging due to post-transcriptional regulation, as it can still reveal the downstream consequences of pGM-CSF signaling even when the protein itself is difficult to detect .

What are the optimal conditions for studying pGM-CSF expression in porcine cells?

For optimal study of pGM-CSF expression in porcine cells, researchers should consider cell type, stimulation conditions, and temporal dynamics. For porcine alveolar macrophages (PAMs), lipopolysaccharide (LPS) stimulation at 0.25 mg/mL provides strong induction of pGM-CSF expression, with peak mRNA levels occurring around 5 hours post-stimulation followed by a gradual decline. For spleen cells, different stimuli produce varying kinetics: LPS yields similar patterns to PAMs, while phytohemagglutinin (PHA) and concanavalin A (ConA) induce more gradual increases with peaks at around 24 hours. RNA isolation should be performed using high-quality reagents like RNAiso Plus, followed by reverse transcription with reliable kits such as PrimeScript RT reagent Kit. For protein detection, a double-antibody sandwich ELISA with mouse monoclonal and rabbit polyclonal antibodies against pGM-CSF offers higher sensitivity than commercial kits. Researchers should always include appropriate time course analyses (0-48 hours) to capture the full expression dynamics, as pGM-CSF levels change significantly over time. Additionally, simultaneous measurement of both mRNA and protein is critical, particularly when studying viral infections that may induce post-transcriptional regulation .

How should experiments be designed to investigate pGM-CSF's effects on PRRSV replication?

Designing experiments to investigate pGM-CSF's effects on PRRSV replication requires careful consideration of multiple factors. A comprehensive experimental design should include both in vitro and in vivo components. For in vitro studies, researchers should use primary porcine alveolar macrophages (PAMs) as they are the natural target cells for PRRSV. Experiments should include: (1) Pre-treatment of PAMs with recombinant pGM-CSF at various concentrations (dose-response) before PRRSV infection; (2) Post-infection treatment to assess therapeutic potential; (3) pGM-CSF knockdown using siRNA to evaluate the role of endogenous pGM-CSF; and (4) Assessment of viral replication through methods like qPCR for viral RNA, immunofluorescence for viral proteins, and titration of infectious virus. For in vivo studies, experimental design should include different PRRSV strains (high pathogenic, classical, and attenuated), measurement of serum pGM-CSF levels at multiple time points, correlation with viral loads and disease parameters, and analysis of downstream signaling in PAMs isolated from infected animals. Controls must include uninfected cells/animals, irrelevant cytokine treatments, and different PRRSV strains to account for strain-specific effects. This comprehensive approach will help reconcile contradictory findings regarding pGM-CSF's role in PRRSV infection .

What controls are essential when analyzing pGM-CSF signaling pathways?

When analyzing pGM-CSF signaling pathways, several essential controls must be incorporated to ensure experimental validity and interpretable results. For in vitro signaling studies, researchers should include: (1) Unstimulated negative controls to establish baseline signaling activity; (2) Positive controls using known activators of relevant pathways (e.g., IFN-γ for STAT1 activation); (3) Dose-response assessments with recombinant pGM-CSF to determine concentration-dependent effects; (4) Time-course experiments to capture transient signaling events; (5) Pathway inhibitor controls using specific chemical inhibitors or siRNAs targeting key signaling molecules; and (6) Species-specificity controls comparing porcine GM-CSF with human or mouse GM-CSF to identify conserved versus species-specific signaling. For downstream gene expression analysis, appropriate housekeeping genes must be validated for the specific experimental conditions. When performing transcriptome analysis, technical replicates are necessary to control for methodological variation, and biological replicates are essential to account for individual variability. Additionally, when studying the effects of viral infection on pGM-CSF signaling, UV-inactivated virus controls can help distinguish between effects requiring viral replication versus those triggered by viral particle recognition .

How can researchers reconcile contradictory data regarding pGM-CSF levels and viral protection?

Reconciling contradictory data regarding pGM-CSF levels and viral protection requires a systematic approach that considers multiple experimental variables. First, researchers should distinguish between in vitro and in vivo findings, as the complex immunological environment in vivo may yield different results than simplified cell culture systems. Second, viral strain differences must be considered—effects observed with PRRSV-1 may differ from those with PRRSV-2, and highly pathogenic strains may interact differently with pGM-CSF than attenuated strains. Third, cell type specificity is crucial—pGM-CSF's effects on porcine alveolar macrophages may differ from its effects on peripheral blood monocytes or dendritic cells. Fourth, temporal dynamics should be analyzed, as pGM-CSF may have different effects at different stages of infection. Fifth, researchers should examine direct versus indirect effects, considering that pGM-CSF may not directly inhibit viral replication but could enhance immune responses that contribute to viral clearance. Finally, methodological differences in pGM-CSF detection and quantification should be evaluated, as different assays have varying sensitivities and specificities. By systematically addressing these variables through carefully designed comparative studies, researchers can develop a more nuanced understanding of pGM-CSF's role in viral protection .

How can a double-antibody sandwich ELISA for pGM-CSF be developed and optimized?

Developing and optimizing a double-antibody sandwich ELISA for pGM-CSF requires careful consideration of antibody selection, assay conditions, and validation procedures. The process begins with generating high-quality antibodies: mouse monoclonal antibodies typically serve as capture antibodies due to their high specificity, while rabbit polyclonal antibodies function as detection antibodies to recognize multiple epitopes. For optimal sensitivity, antibody pairs should recognize non-overlapping epitopes on the pGM-CSF molecule. The assay development process involves coating microplates with the capture antibody at optimized concentration, blocking non-specific binding sites, adding samples or standards, adding the detection antibody, and finally adding an enzyme-conjugated secondary antibody followed by substrate. Critical optimization steps include determining the optimal antibody concentrations, incubation times and temperatures, washing procedures, and blocking reagents. Validation should assess specificity (cross-reactivity testing), sensitivity (lower limit of detection), precision (intra- and inter-assay variation), accuracy (spike recovery tests), and linearity of dilution. Based on research findings, such optimized in-house ELISA systems can achieve higher sensitivity than commercial kits, making them valuable tools for evaluating pGM-CSF levels in both cell culture supernatants and serum samples from in vivo experiments .

What approaches can be used to study post-transcriptional regulation of pGM-CSF?

Studying post-transcriptional regulation of pGM-CSF, particularly during viral infections like PRRSV, requires multiple complementary approaches. Researchers can employ polysome profiling to analyze mRNA association with ribosomes, providing insights into translation efficiency. This technique separates mRNAs based on the number of associated ribosomes through sucrose gradient centrifugation, followed by fraction collection and RT-qPCR analysis of pGM-CSF mRNA distribution. RNA stability assays using actinomycin D to block transcription can determine whether accelerated mRNA degradation contributes to reduced protein levels. RNA immunoprecipitation (RIP) can identify RNA-binding proteins that interact with pGM-CSF mRNA and potentially regulate its translation or stability. For viral interference mechanisms, reporter constructs containing the pGM-CSF 5' and 3' untranslated regions fused to luciferase can reveal whether these regulatory regions are targeted during viral infection. Additionally, CRISPR-Cas9 screening can identify host factors required for post-transcriptional regulation. Comparative analysis between different PRRSV strains can determine whether post-transcriptional suppression correlates with virulence. These approaches, used in combination, can elucidate the mechanisms by which PRRSV infection induces post-transcriptional inhibition of pGM-CSF production .

How can RNA-seq data be effectively analyzed to understand pGM-CSF-regulated gene networks?

Effective analysis of RNA-seq data to understand pGM-CSF-regulated gene networks requires a comprehensive bioinformatics pipeline with special considerations for immunological context. The analysis workflow should begin with quality control of raw sequencing data (FastQC), followed by read trimming/filtering, alignment to the porcine reference genome, and quantification of gene expression. For differential expression analysis, tools like DESeq2 or edgeR should be employed with appropriate statistical thresholds (typically adjusted p-value < 0.05 and fold-change > 1.5). Since pGM-CSF influences macrophage polarization, specific attention should be paid to M1 (pro-inflammatory) versus M2 (anti-inflammatory) gene signatures. Pathway analysis using tools like GSEA, IPA, or KEGG enrichment can identify biological processes and signaling networks activated by pGM-CSF. Time-course RNA-seq data should be analyzed using specialized tools like maSigPro or ImpulseDE2 that can identify temporal patterns in gene expression. Integration of transcriptomic data with pGM-CSF protein levels and functional outcomes (e.g., viral loads, cytokine production) can reveal correlations between gene expression patterns and biological effects. Network analysis tools like WGCNA can identify co-expressed gene modules associated with pGM-CSF signaling. Finally, validation of key findings using RT-qPCR, protein analysis, or functional assays is essential to confirm the biological relevance of identified signature genes or pathways .

How can pGM-CSF be utilized as an immune adjuvant in PRRSV vaccines?

Porcine GM-CSF holds significant potential as an immune adjuvant for PRRSV vaccines based on its immunomodulatory properties. Research has demonstrated that incorporating pGM-CSF into vaccine formulations can enhance both humoral and cell-mediated immune responses. Piglets inoculated with recombinant live attenuated PRRSV vaccine (MLV) carrying the pGM-CSF gene exhibited lower viremia, reduced lung lesions, and higher serum levels of IFN-γ compared to animals receiving MLV alone. Similarly, immunization with adenovirus-vectored PRRSV-GP3/GP5 co-expressing pGM-CSF induced significantly higher PRRSV-specific neutralizing antibodies and increased production of both IFN-γ (Th1-associated) and IL-4 (Th2-associated) cytokines. For practical applications, pGM-CSF can be incorporated into vaccines through several approaches: genetic fusion with viral antigens, co-expression from viral vectors, formulation as a protein adjuvant, or delivery via plasmid DNA. The optimal delivery method depends on the vaccine platform (live attenuated, subunit, vectored, etc.) and desired immune response profile. Future vaccine development should focus on optimizing pGM-CSF dose, timing, and delivery method to maximize protective immunity while minimizing potential adverse effects associated with cytokine administration .

What are the potential therapeutic applications of His-tagged pGM-CSF in porcine disease models?

His-tagged pGM-CSF offers several potential therapeutic applications in porcine disease models beyond its adjuvant properties. The His-tag provides advantages for purification, detection, and potentially extended half-life, making this form particularly valuable for research and therapeutic development. In PRRSV infection models, His-tagged pGM-CSF could be evaluated as an immunomodulatory therapy to enhance macrophage activation and antiviral responses, particularly when administered alongside antiviral treatments like neutralizing antibodies. Transcriptome studies suggest that pGM-CSF promotes an M1-like macrophage phenotype that may contribute to viral clearance. For bacterial respiratory infections, His-tagged pGM-CSF could enhance neutrophil and macrophage function, improving bacterial clearance. In wound healing and tissue repair models, it could promote monocyte recruitment and differentiation. The His-tag allows for tracking the biodistribution and pharmacokinetics of administered pGM-CSF, providing crucial data for therapeutic development. Additionally, His-tagged pGM-CSF could serve as a research tool for pulling down receptor complexes and identifying binding partners, advancing our understanding of species-specific signaling pathways. As production systems and delivery methods improve, His-tagged pGM-CSF has potential applications in various porcine disease models where immune enhancement or modulation is beneficial .

How might future research resolve the contradictory findings regarding pGM-CSF's role in viral infections?

Future research to resolve contradictory findings regarding pGM-CSF's role in viral infections should adopt a multifaceted approach addressing key knowledge gaps. Researchers should conduct comprehensive studies comparing multiple PRRSV strains (PRRSV-1 vs. PRRSV-2, high-pathogenic vs. attenuated) in the same experimental system to determine whether strain-specific factors influence pGM-CSF's effects. Parallel in vitro and in vivo experiments are essential to understand how the complex immunological environment in vivo might lead to different outcomes than simplified cell culture systems. Single-cell RNA sequencing of porcine lung cells during infection could reveal cell type-specific responses to pGM-CSF that may explain contradictory findings. Mechanistic studies exploring the post-transcriptional regulation of pGM-CSF during viral infection would clarify how viruses modulate its expression. Knock-in or knockout pig models with altered pGM-CSF expression or signaling would provide definitive evidence of its role during infection. Longitudinal studies examining pGM-CSF levels and effects at different infection stages could resolve temporal contradictions. Finally, standardization of detection methods, quantification approaches, and experimental protocols across research groups would reduce technical variables that may contribute to contradictory results. This comprehensive approach would provide a more nuanced understanding of pGM-CSF's context-dependent roles in viral infections .

What are the key technical challenges in working with His-tagged pGM-CSF and how can they be overcome?

Working with His-tagged pGM-CSF presents several technical challenges that researchers must address for successful experimentation. First, bacterial expression systems often produce insoluble protein in inclusion bodies, requiring optimization of expression conditions (lower temperature, reduced inducer concentration) or refolding protocols to obtain active protein. Second, endotoxin contamination from E. coli expression systems can confound immunological experiments; this requires rigorous endotoxin removal using polymyxin B columns or specific endotoxin removal resins, with validation by LAL assay. Third, proper protein folding and biological activity must be verified, as the His-tag may occasionally interfere with protein structure or function; activity assays comparing tagged and untagged versions can address this concern. Fourth, protein stability during storage presents challenges; optimizing buffer conditions (pH, salt concentration, glycerol percentage) and storage temperature (-80°C with flash freezing) can maximize shelf-life. Fifth, batch-to-batch variation must be minimized through standardized production protocols and quality control measures. Finally, the development of detection reagents specific for pGM-CSF rather than the His-tag is essential for accurate quantification in biological samples. Overcoming these challenges requires systematic optimization of expression, purification, and storage conditions, along with rigorous quality control measures to ensure consistent, biologically active protein for research applications .

Product Science Overview

Porcine GM-CSF

Porcine GM-CSF (pGM-CSF) is specifically derived from pigs and has been studied for its unique expression patterns and functional roles in porcine immune cells . The expression of pGM-CSF can be induced by various stimulants such as lipopolysaccharide (LPS), phytohemagglutinin (PHA), and concanavalin A (ConA) . These stimulants trigger the production of pGM-CSF in immune cells like alveolar macrophages and spleen cells .

Recombinant GM-CSF

Recombinant GM-CSF refers to the GM-CSF protein that has been genetically engineered and produced using recombinant DNA technology. This involves cloning the GM-CSF gene and expressing it in a suitable host system, such as bacteria or yeast, to produce the protein in large quantities . The recombinant form of GM-CSF is used in various research and therapeutic applications due to its ability to stimulate the immune system.

His Tag

A His Tag (Histidine Tag) is a sequence of histidine residues added to proteins to facilitate their purification and detection. The His Tag binds strongly to nickel or cobalt ions, allowing the tagged protein to be easily isolated using affinity chromatography . In the case of recombinant pGM-CSF, the His Tag is added to simplify the purification process and ensure the protein’s purity for research and clinical use .

Applications and Research

Recombinant pGM-CSF with a His Tag has been utilized in various studies to understand its immunological functions and potential therapeutic applications. For instance, it has been used to enhance the immunogenicity and antiviral protection of vaccines in piglets . Additionally, the recombinant form of pGM-CSF has shown promise as an adjuvant in swine immunization and viral challenge studies .

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.