Property | Specification | Method of Determination |
---|---|---|
Molecular Weight | ~6.8 kDa | Theoretical calculation |
Expression Region | Amino acids 46-107 | Protein sequencing |
Purity | >97% | SDS-PAGE and HPLC analysis |
Endotoxin Level | <1.0 EU/μg | LAL method |
Form | Lyophilized powder | Physical observation |
Buffer Composition | 0.2 μm filtered PBS, pH 7.4 | Buffer analysis |
Protein Length | Partial | Sequence analysis |
Tag Configuration | Tag-Free | Expression design |
The biological significance of Recombinant Rat CXC chemokine RTCK1 extends across multiple immune regulatory pathways. In vivo, PPBP is primarily expressed in monocytes and macrophages, particularly following exposure to bacterial endotoxins, and is released from the alpha-granules of platelets upon activation . The recombinant form maintains full biological activity comparable to the standard protein, demonstrating chemotactic properties for both fibroblasts and neutrophils .
Activity assays reveal that this recombinant protein exhibits biological effectiveness within a concentration range of 0.1-1.0 ng/ml, specifically when tested using human CXCR2 transfected murine BaF3 cells in chemotaxis bioassays . This specific interaction with the CXCR2 receptor underscores its role in immune signaling and inflammatory cascades . Beyond simple chemotaxis, PPBP participates in a diverse array of cellular processes including mitogenesis, DNA synthesis, extracellular matrix synthesis, glycolysis, and plasminogen activator synthesis .
Within rat models, this chemokine represents an important analytical target for understanding immune regulation and inflammatory processes. Its presence in various immunological pathways makes it valuable for studying conditions involving neutrophil recruitment and activation . The precise activity profile of the recombinant protein makes it suitable for in vitro studies seeking to elucidate these mechanisms in controlled experimental conditions.
The production of Recombinant Rat CXC chemokine RTCK1 protein involves sophisticated expression in E. coli systems, followed by rigorous purification and quality control measures . The resulting protein demonstrates exceptional purity exceeding 97% as verified through both SDS-PAGE and High-Performance Liquid Chromatography (HPLC) analyses . This high level of purity is essential for ensuring experimental reproducibility and eliminating potential confounding factors in research applications.
Endotoxin control represents another critical quality parameter, with levels maintained below 1.0 EU/μg as determined by the Limulus Amebocyte Lysate (LAL) method . This stringent endotoxin limit is particularly important for immunological research, where even trace amounts of bacterial endotoxins could trigger unwanted immune responses and compromise experimental outcomes.
The recombinant protein is typically supplied as a lyophilized powder, providing optimal stability during shipping and storage . This formulation is prepared by lyophilizing the protein from a 0.2 μm filtered phosphate-buffered saline (PBS) solution at pH 7.4, ensuring both purity and physiological compatibility .
Recombinant Rat CXC chemokine RTCK1 protein serves as a valuable tool in immunological research, particularly in studies focused on neutrophil activation, chemotaxis, and inflammatory responses. Its well-characterized activity profile enables researchers to investigate specific immune signaling pathways with high precision.
In rat model systems, this recombinant protein can be used to examine the effects of CXCL7/PPBP on various cellular processes including migration, proliferation, and activation of immune cells . The protein's interaction with the CXCR2 receptor makes it particularly useful for research focused on this signaling axis, including studies on inflammatory disorders, wound healing, and immune cell recruitment .
The protein is compatible with various experimental sample types, including cell culture supernatants, cell lysates, plasma, serum, and tissue lysates, providing flexibility for different research approaches . Its application in chemokine arrays allows for multiplex analysis of immune responses, offering insights into complex inflammatory cascades and regulatory networks .
For immunological research focusing on rat models, this recombinant protein represents one component of a broader analytical toolkit. Comprehensive chemokine arrays incorporating this protein alongside other inflammatory mediators can provide a more complete picture of immune regulation in various experimental conditions .
Within the broader context of chemokine biology, Recombinant Rat CXC chemokine RTCK1 protein (PPBP/CXCL7) exists alongside several other significant CXC chemokines in rat systems, including CINC-1 (CXCL1), CINC-2, CINC-3, LIX, and MIG (CXCL9) . Together, these chemokines form a complex regulatory network governing immune cell recruitment and activation in response to various stimuli.
The specificity of CXCL7 for the CXCR2 receptor distinguishes it functionally from other chemokines that bind different receptors . This receptor specificity determines the cellular targets and downstream effects of the chemokine, contributing to its unique role in immune regulation.
Understanding the position of CXCL7/PPBP within this chemokine network provides valuable context for interpreting research findings and designing experimental approaches. The availability of high-quality recombinant proteins enables comparative studies examining the distinct and overlapping roles of different chemokines in immune regulation and inflammatory processes.
RTCK1 protein (Ppbp), also known as Cxcl7, is a proinflammatory chemokine expressed in rats with a molecular weight of approximately 6.8 kDa . The protein is structurally characterized by its CXC motif and functions primarily through binding to CXCR2 receptors. In biological systems, this protein plays a critical role in neutrophil migration and activation during inflammatory responses . The protein mediates its effects by promoting chemotaxis, extravasation, respiratory burst, and degranulation of neutrophils . Additionally, it can induce T cells to produce proinflammatory IL-17, contributing to the inflammatory cascade . The recombinant form typically encompasses the expression region 46-107aa with the sequence "IELRCRCTNT LSGIPLNSIS RVNVFRPGAH CDNVEVIATL KNGKEVCLDP TAPMIKKIVK KI" .
RTCK1/Ppbp belongs to the CXC chemokine family which includes related proteins like CXCL1/GRO alpha/KC/CINC-1 . While sharing structural similarities with other CXC chemokines, RTCK1 has specific functional characteristics. Comparative analysis shows that rat CXCL1, a related chemokine, shares 67% amino acid sequence identity with human CXCL1 and 92% with mouse CXCL1 . Similar homology patterns may exist for RTCK1/Ppbp, explaining cross-species reactivity in certain experimental systems. Functionally, like other CXC chemokines, RTCK1 can associate into bioactive dimers that primarily signal through CXCR2/IL-8 RB receptors, though it can also bind with lower affinity to CXCR1/IL-8 RA . The evolutionary conservation of these proteins highlights their biological importance across mammalian species.
When working with the partial active form of RTCK1 protein, researchers should consider several critical factors. First, the partial nature of the recombinant protein (typically encompassing amino acids 46-107) may affect certain structural elements compared to the full-length protein . The expression system used (E. coli) means the protein lacks eukaryotic post-translational modifications, which may influence certain functional studies . Researchers should validate biological activity using appropriate assays, such as chemotaxis bioassays with CXCR2-transfected murine BaF3 cells, where the active concentration range is typically 0.1-1.0 ng/mL . Proper reconstitution is essential - the lyophilized protein should be reconstituted in deionized sterile water to a concentration of 0.1-1.0 mg/mL, preferably with 5-50% glycerol added for storage stability . Avoid repeated freeze-thaw cycles as these can significantly diminish protein activity.
Establishing optimal reconstitution and storage conditions is critical for maintaining RTCK1 protein activity. Begin by briefly centrifuging the lyophilized protein vial before opening to bring contents to the bottom . Reconstitute the protein in deionized sterile water to a concentration between 0.1-1.0 mg/mL under aseptic conditions . For long-term storage stability, add glycerol to a final concentration of 5-50% (with 50% being standard practice for many laboratories) . Aliquot the reconstituted protein into smaller volumes to minimize freeze-thaw cycles and store at -20°C to -80°C for long-term preservation . For short-term use, working aliquots can be stored at 4°C for up to one week . Researchers should validate protein activity after reconstitution using functional assays specific to chemokines, such as chemotaxis assays with CXCR2-expressing cells. Regular quality control testing of stored aliquots is recommended, particularly for experiments requiring precise quantification of biological activity.
Testing RTCK1 protein activity in vitro requires specific methodologies and appropriate cellular models. The gold standard for RTCK1 activity assessment is the chemotaxis bioassay using CXCR2-transfected murine BaF3 pro-B cells, with functional activity typically observed in the concentration range of 0.1-1.0 ng/mL . This assay measures the protein's ability to attract responsive cells across a membrane, quantifying migration as a function of concentration. Alternative cell models include human or rat neutrophils, which naturally express CXCR2 receptors. Calcium flux assays can also be employed to measure immediate receptor activation upon RTCK1 binding. For molecular interaction studies, binding assays using cells expressing CXCR1 or CXCR2 can determine receptor specificity and binding kinetics . When designing these experiments, include appropriate positive controls (like commercial CXCL1/CINC-1 with established activity) and negative controls (non-chemotactic proteins or buffer alone) to validate assay performance.
Distinguishing between endogenous and recombinant RTCK1 protein in experimental systems requires strategic approaches. One effective method is to utilize tagged recombinant versions of the protein, though researchers should note that the product described is tag-free . Alternative approaches include using species-specific antibodies when working in cross-species systems (e.g., rat recombinant protein in human or mouse cell lines). Researchers can also leverage the known molecular weight differences - the recombinant partial protein has a molecular weight of 6.8 kDa, which may differ from the endogenous full-length protein . For more precise discrimination, mass spectrometry can identify unique peptide signatures specific to the recombinant version. When conducting in vitro experiments, establishing proper baseline measurements in control samples is crucial for quantifying the contribution of exogenously added recombinant protein versus endogenous production. Finally, knockdown or knockout models of the endogenous protein can create clean systems for studying recombinant protein function.
The expression efficiency of recombinant RTCK1 protein is significantly influenced by mRNA accessibility, particularly around the translation initiation site. Research shows that the accessibility of translation initiation sites modeled using mRNA base-unpairing across the Boltzmann's ensemble is a powerful predictor of recombinant protein expression success . The region spanning positions -24 to +24 relative to the initiation codon has been identified as particularly critical, with an area under the receiver operating characteristic curve (AUC) of approximately 0.70 in predicting expression outcomes . To optimize expression, researchers can employ synonymous codon substitutions within the first nine codons of the mRNA sequence without altering the amino acid sequence . Tools like TIsigner (Translation Initiation coding region designer) can be utilized to predict these modifications . This approach is cost-effective as it requires only PCR-based modifications rather than full-length gene synthesis . Implementing these strategies can significantly improve the yield of recombinant RTCK1 protein from E. coli expression systems.
E. coli-derived recombinant RTCK1 protein fundamentally differs from native rat RTCK1 in its post-translational modification (PTM) profile, which can significantly impact experimental outcomes. As a prokaryotic expression system, E. coli lacks the cellular machinery for eukaryotic modifications such as glycosylation, phosphorylation, and specific proteolytic processing that may occur in native rat cells . These differences can affect protein folding, stability, receptor binding affinity, and biological half-life. In chemotaxis assays, while the E. coli-derived protein remains biologically active (functional at 0.1-1.0 ng/mL in CXCR2-transfected BaF3 cells), its potency may differ from the native form . For studies where PTMs are critical, researchers should consider alternative expression systems (mammalian, insect, or yeast cells) or complementary approaches using native protein purified from rat tissues. When interpreting results, particularly in translational research, these limitations should be explicitly acknowledged. Comparative studies examining activity differences between bacterial-derived and eukaryotic-expressed RTCK1 can provide valuable insights into the functional relevance of specific PTMs.
Studying RTCK1 protein dimerization and its impact on receptor signaling requires sophisticated methodological approaches. Like other CXC chemokines, RTCK1 can associate into bioactive dimers that influence receptor binding dynamics . To investigate dimerization, researchers can employ size exclusion chromatography to separate monomeric and dimeric forms under physiological conditions. Analytical ultracentrifugation and dynamic light scattering provide additional quantitative measurements of oligomerization states. For structural insights, cross-linking studies followed by mass spectrometry can identify specific residues involved in dimer interfaces. To assess functional consequences, comparative receptor binding assays using surface plasmon resonance can determine whether monomeric and dimeric forms exhibit different binding kinetics to CXCR1 and CXCR2 receptors . Downstream signaling can be evaluated through phosphorylation studies of key pathway components (e.g., ERK1/2, Akt) and calcium mobilization assays in receptor-expressing cells. Creating mutant versions of RTCK1 with altered dimerization potential can further elucidate structure-function relationships in receptor activation and signaling cascade initiation.
Poor solubility or aggregation of recombinant RTCK1 protein represents a common technical challenge that can be addressed through multiple strategies. First, optimize the reconstitution process by using appropriate buffers - while PBS (pH 7.4) is standard , testing buffer systems with different pH values (6.8-8.0) or ionic strengths may improve solubility. Add solubilizing agents such as low concentrations of non-ionic detergents (0.01-0.05% Tween-20) or carrier proteins (0.1% BSA) to prevent aggregation, particularly for dilute solutions. Sonication with short pulses or filtration through a 0.22 μm filter can help disperse existing aggregates. Temperature management is crucial - perform reconstitution at room temperature or 4°C rather than directly from frozen, and allow the protein to equilibrate fully before use. For persistent aggregation issues, consider testing different reconstitution methods such as step-wise dilution from a concentrated stock. Finally, centrifugation at 10,000g for 10 minutes before use can remove any insoluble aggregates. Document successful approaches carefully, as optimal conditions may vary between different protein preparations.
Inconsistent results in chemotaxis assays using recombinant RTCK1 protein can stem from multiple sources that require systematic troubleshooting. Begin by validating protein activity using positive controls like commercially available CXCL1/CINC-1 with established chemotactic properties . Ensure the protein concentration falls within the optimal range (0.1-1.0 ng/mL for CXCR2-transfected BaF3 cells) , as both too low and too high concentrations can yield unreliable results. Verify receptor expression levels on target cells through flow cytometry or Western blotting, as receptor downregulation may occur after repeated passaging. Standardize cell culture conditions, ensuring consistent cell density, passage number, and serum starvation protocols. The chemotaxis chamber configuration is critical - check for air bubbles, membrane integrity, and proper assembly. Incubation time and temperature should be precisely controlled, as even minor variations can significantly impact cell migration. For quantification, establish objective counting methods, ideally using automated imaging systems to reduce observer bias. Finally, consider the formation of chemokine gradients - improper gradient formation can lead to random migration rather than directed chemotaxis.
Incorporating RTCK1 protein into tissue-engineered models offers powerful insights into inflammatory processes but requires methodological refinement. Begin by determining the optimal delivery method - options include direct addition to culture media, controlled-release from biodegradable microparticles, or genetically modified cells that secrete the protein upon induction. For three-dimensional tissue models, establish concentration gradients that mimic physiological conditions using microfluidic systems with independent chambers. The protein stability within the tissue microenvironment should be assessed through immunodetection at various timepoints, recognizing that matrix components and cellular proteases may affect protein half-life. Validation of RTCK1 activity within the tissue context is essential - measure neutrophil infiltration patterns, expression of inflammatory mediators, and activation of CXCR2-dependent signaling pathways. For temporal control, consider photoactivatable or enzyme-responsive protein conjugates that enable spatially precise activation. When designing these experiments, include appropriate controls including non-functional protein variants and CXCR2 antagonists to confirm specificity. This approach enables more physiologically relevant studies of RTCK1 function compared to traditional two-dimensional cell cultures.
Designing effective CRISPR/Cas9 experiments for RTCK1/Ppbp studies requires careful consideration of multiple factors. Begin by identifying suitable target sites within the gene locus - depending on experimental goals, targets may include the coding region (for complete knockout), promoter elements (for expression modulation), or specific functional domains (for protein variant generation). Bioinformatic tools should be employed to design guide RNAs with high on-target efficiency and minimal off-target effects, with particular attention to potential cross-reactivity with related CXC chemokine genes given their sequence similarities. For validation, design PCR primers that can discriminate between wild-type and edited alleles, and plan sequencing strategies to confirm precise edits. Consider the cellular context carefully - primary rat cells, immortalized cell lines, or in vivo editing each present different efficiency challenges and physiological relevance. For functional studies, establish appropriate readouts including chemokine expression levels, secretion patterns, and downstream inflammatory responses. When analyzing phenotypes, account for potential compensatory mechanisms involving other chemokines, as redundancy within the CXC family may mask certain functional deficits after RTCK1/Ppbp modification.
Emerging technologies are poised to transform our understanding of RTCK1 protein interactions in complex disease models. Single-cell technologies, including single-cell RNA sequencing and CyTOF mass cytometry, can now map RTCK1 production and receptor expression across heterogeneous cell populations within inflamed tissues, revealing previously unrecognized cellular sources and targets. Proximity labeling approaches such as BioID or APEX2 enable in situ identification of transient RTCK1 binding partners in living cells, potentially uncovering novel signaling complexes beyond canonical receptor interactions . Advances in cryo-electron microscopy now permit structural determination of chemokine-receptor complexes at near-atomic resolution, offering unprecedented insights into binding specificity determinants. Organ-on-chip platforms incorporating primary rat cells can model RTCK1 function within physiologically relevant tissue architectures and flow conditions. For in vivo studies, intravital microscopy combined with fluorescently labeled RTCK1 allows real-time visualization of chemokine gradients and cellular responses. Looking forward, CRISPR-based epigenetic modifiers will enable precise manipulation of RTCK1 expression patterns, while computational approaches integrating multi-omics data will situate RTCK1 within broader inflammatory networks, advancing our understanding of its role in complex disease pathogenesis.