GRO-Alpha/MGSA Human Recombinant (CXCL1) with His Tag is a small cytokine belonging to the CXC chemokine family. It is known by several synonyms in scientific literature, including Growth-regulated protein alpha, Melanoma growth stimulatory activity (MGSA), Neutrophil-activating protein 3 (NAP-3), GRO-alpha (1-73), chemokine (C-X-C motif) ligand 1, GRO1, GROa, SCYB1, and MGSA alpha . The His Tag refers to a polyhistidine tag commonly added to recombinant proteins to facilitate purification and detection in experimental contexts. The recombinant version maintains the biological functions of the native protein while offering enhanced utility for laboratory research applications .
CXCL1 serves multiple important biological functions that make it a significant target for research. It is secreted by human melanoma cells and has established mitogenic properties, being implicated in melanoma pathogenesis. The protein is naturally expressed by macrophages, neutrophils, and epithelial cells throughout the body . One of its primary functions is serving as a neutrophil chemoattractant, playing a crucial role in immune cell recruitment during inflammatory responses. Additionally, CXCL1 has been identified as an important factor in spinal cord development, specifically by inhibiting the migration of oligodendrocyte precursor cells . This multifunctionality makes CXCL1 relevant to research in oncology, immunology, and neurodevelopment fields.
To maintain optimal activity of GRO-Alpha/CXCL1 preparations, researchers should adhere to strict storage and handling protocols. The lyophilized protein should be stored at -20°C and protected from light. Once reconstituted, the protein should be aliquoted to avoid repeated freeze-thaw cycles and stored at -80°C for long-term preservation. When working with the protein, maintain sterile conditions and use low-protein binding tubes to prevent adhesion and loss of material. The reconstitution buffer should typically be phosphate-buffered saline (PBS) containing a small percentage (0.1%) of human or bovine serum albumin as a carrier protein. Always validate protein activity after extended storage periods using appropriate bioassays that measure chemotactic activity or receptor binding. Researchers should note that His-tagged versions may exhibit slightly different stability profiles compared to untagged versions, necessitating preliminary stability testing in specific experimental conditions.
Various experimental models have proven effective for studying CXCL1 functions across different research contexts. For genetic and molecular studies, yeast cells provide a simplified eukaryotic system where specific aspects of protein function can be isolated . Zebrafish models have demonstrated particular utility for studying developmental roles of CXCL1, offering advantages of transparency during development and genetic manipulability . For cancer-related research, human melanoma cell lines are frequently employed given CXCL1's established role in melanoma pathogenesis . In immunological studies, neutrophil migration assays using Boyden chambers or microfluidic devices provide direct functional readouts of CXCL1 chemotactic activity. Mouse models with selective knockout or overexpression of CXCL1 are valuable for in vivo studies of inflammatory conditions, wound healing, and tumor growth. When selecting an experimental model, researchers should consider the specific CXCL1 function being investigated and whether the model appropriately expresses the cognate receptors for human CXCL1.
Purification of His-tagged CXCL1 typically employs immobilized metal affinity chromatography (IMAC) as the primary isolation method. Most protocols begin with bacterial or mammalian expression systems, followed by cell lysis under native or denaturing conditions depending on whether the protein forms inclusion bodies. For IMAC purification, nickel or cobalt resins are commonly used, with initial binding in a buffer containing 10-20 mM imidazole to reduce non-specific binding, followed by washing steps and elution with 250-500 mM imidazole. Size exclusion chromatography is recommended as a secondary purification step to remove aggregates and ensure monomeric protein preparation. For enhanced purity, researchers may implement ion exchange chromatography as an additional step. Throughout the purification process, SDS-PAGE and Western blotting with anti-His antibodies should be used to track protein presence and purity. When designing a purification protocol, researchers should consider that CXCL1's small size (approximately 8-11 kDa without the tag) may influence its behavior during purification, potentially necessitating specialized conditions for optimal recovery and activity preservation.
Validating bioactivity of purified CXCL1 requires multiple complementary approaches. The neutrophil chemotaxis assay serves as the gold standard functional test, measuring the protein's ability to induce directional migration of neutrophils in a concentration-dependent manner. Researchers should perform receptor binding assays using cells expressing CXCR2 (the primary receptor for CXCL1) to confirm specific binding activity, typically through radioligand competition or surface plasmon resonance techniques. Calcium flux assays provide a rapid assessment of signaling capability, as CXCL1 binding to its receptor triggers intracellular calcium release that can be measured with fluorescent indicators. For confirming biological relevance, endothelial cell tube formation assays can assess angiogenic activity, while scratch wound healing assays can measure effects on cell migration and proliferation. Each validation method should include appropriate positive controls (commercial CXCL1) and negative controls (buffer only or heat-inactivated protein) to ensure result reliability. Activity should be expressed as EC50 values (effective concentration producing 50% of maximum response) to enable meaningful comparisons between different preparations.
Studying the structure-function relationship of CXCL1 presents several significant challenges. The protein's small size (8-11 kDa) and tendency to form homodimers at higher concentrations complicate structural analyses and functional interpretations. X-ray crystallography and NMR studies have revealed that CXCL1 undergoes conformational changes upon receptor binding, but capturing these transient states remains technically challenging. The presence of a His-tag, while useful for purification, may influence structural dynamics, requiring careful control experiments to distinguish native from artifact behaviors. Post-translational modifications, particularly N-terminal processing, significantly impact CXCL1 activity, but producing homogeneously modified recombinant preparations is difficult. Additionally, CXCL1 interactions with glycosaminoglycans (GAGs) in the extracellular matrix are critical for in vivo function but are often overlooked in simplified in vitro systems. Researchers addressing these challenges should consider employing hydrogen-deuterium exchange mass spectrometry (HDX-MS) to capture dynamic conformational changes, use site-directed mutagenesis to probe functional domains systematically, and develop physiologically relevant 3D culture systems incorporating extracellular matrix components to better reflect in vivo conditions.
CXCL1 signaling intersects with multiple chemokine pathways in complex disease models through several mechanisms. In inflammatory conditions, CXCL1 works synergistically with other ELR+ CXC chemokines (CXCL2, CXCL3, CXCL8) that share the CXCR2 receptor, creating functional redundancy that complicates targeted interventions. Cross-talk between CXCL1 and CCL2 pathways has been documented in tumor microenvironments, where CXCL1-recruited neutrophils subsequently alter monocyte recruitment through CCL2 modulation. The presence of atypical chemokine receptors (ACKRs) adds another layer of complexity by scavenging CXCL1 and altering local concentration gradients without initiating classical signaling. In neurodevelopmental contexts, CXCL1 signaling interacts with PDGF and Notch pathways to regulate oligodendrocyte precursor cell migration and differentiation, relationships that remain incompletely characterized . To effectively study these interactions, researchers should implement multi-omics approaches that capture changes across the chemokine network, develop multiplexed imaging techniques to visualize multiple chemokine gradients simultaneously, and utilize conditional genetic models that allow temporal control over specific pathway components. Single-cell technologies are particularly valuable for deconvoluting heterogeneous responses across different cell populations within complex tissues.
When designing experiments to study CXCL1 in cancer research, several critical considerations must be addressed. The heterogeneity of CXCL1 expression across different cancer types necessitates careful selection of appropriate cell lines or patient-derived samples, with melanoma models being particularly relevant given CXCL1's established role in melanoma pathogenesis . Researchers should account for the tumor microenvironment, as stromal cells often contribute significantly to CXCL1 production, making co-culture systems or 3D organoids more physiologically relevant than cancer cells in isolation. The dual nature of CXCL1 in promoting both tumor growth and anti-tumor immunity requires experimental designs that can distinguish these opposing effects, potentially through selective depletion of specific immune cell populations. Concentration-dependent effects are crucial to consider, as CXCL1 demonstrates different biological activities at varying concentrations. Time-course experiments are essential to capture the dynamic nature of CXCL1 signaling during tumor progression. When translating findings toward therapeutic applications, researchers should incorporate pharmacokinetic/pharmacodynamic modeling to address challenges in targeting a protein with multiple physiological roles. Finally, combinatorial approaches testing CXCL1 interventions alongside standard cancer therapies will provide more clinically relevant insights than studying CXCL1 modulation in isolation.
GRO-Alpha/CXCL1 research offers significant potential for therapeutic development in inflammatory disorders through multiple mechanisms. As a potent neutrophil chemoattractant, CXCL1 represents a promising target for conditions characterized by neutrophilic inflammation, including inflammatory bowel disease, rheumatoid arthritis, and acute lung injury. Inhibition strategies may include neutralizing antibodies, small molecule receptor antagonists targeting CXCR2, or RNA interference approaches to downregulate CXCL1 production. Conversely, controlled delivery of recombinant CXCL1 might enhance immune responses in immunodeficiency conditions or promote tissue repair through controlled neutrophil recruitment. The development of modified CXCL1 variants with altered receptor binding profiles could enable more precise modulation of specific inflammatory pathways while minimizing off-target effects. When designing therapeutics targeting the CXCL1 pathway, researchers must carefully consider the protein's context-dependent roles and potential for compensatory upregulation of related chemokines. Biomarker development represents another valuable application, as CXCL1 levels correlate with disease activity in several inflammatory conditions and could guide personalized treatment approaches. For maximum translational impact, researchers should establish clear connections between preclinical models and human disease mechanisms through parallel studies in patient samples and animal models.
Recent research suggests CXCL1 may have unexpected connections to genetic disorders related to protein synthesis, opening new avenues for investigation. While not directly implicated in the genetic disorder described in search result , the research methodologies using yeast cells and zebrafish to study protein synthesis disorders provide a valuable framework for investigating potential CXCL1 involvement in similar conditions. Protein synthesis is fundamental to CXCL1 expression and function, with proper folding and post-translational modifications being critical for bioactivity. Genetic disorders affecting the translation machinery could potentially alter CXCL1 production or processing, particularly relevant given that CXCL1 contains proline residues that are notoriously difficult for ribosomes to incorporate efficiently. Researchers investigating these connections should consider implementing ribosome profiling techniques to assess translational efficiency of CXCL1 mRNA in disease models, proteomic approaches to identify abnormal CXCL1 processing or modifications, and functional assays to determine whether altered CXCL1 contributes to disease phenotypes. The therapeutic approach mentioned in search result involving spermidine supplementation for protein synthesis disorders suggests that nutritional interventions affecting translation might indirectly modulate CXCL1 biology, representing a potential therapeutic avenue worth exploring in conditions where CXCL1 dysregulation contributes to pathology.
CXCL1 research and neurodevelopmental studies intersect in several significant ways that present valuable research opportunities. CXCL1 plays a documented role in spinal cord development by inhibiting the migration of oligodendrocyte precursor cells, suggesting important functions in myelination processes . This connection positions CXCL1 as a potential target for investigation in demyelinating disorders and developmental myelin abnormalities. Beyond oligodendrocyte regulation, emerging evidence suggests CXCL1 may influence neural progenitor cell proliferation and differentiation, though these mechanisms remain incompletely characterized. The protein's expression by activated microglia during neuroinflammation creates another intersection with neurodevelopmental processes, particularly in conditions where early-life inflammation affects brain development. Researchers exploring these connections should implement lineage tracing techniques to track neural cell development in the presence of CXCL1 modulators, utilize brain organoids to model CXCL1 functions in human neurodevelopment, and employ conditional knockout approaches to determine timing-specific roles of CXCL1 signaling in neural circuit formation. Integration with other neurodevelopmental signaling pathways, particularly Notch and Wnt, which are known to interact with chemokine signaling, represents another productive research direction. Translational applications might include developing CXCL1-based interventions to promote myelin repair in conditions like multiple sclerosis or exploring CXCL1's potential as a biomarker for neuroinflammatory processes in neurodevelopmental disorders.
Reproducibility challenges with recombinant CXCL1 stem from several factors that researchers must systematically address. Batch-to-batch variation represents a primary concern, particularly regarding protein folding, aggregation state, and post-translational modifications that affect bioactivity. To mitigate this, researchers should implement rigorous quality control protocols including SDS-PAGE, mass spectrometry, and circular dichroism to verify protein integrity and conformation across batches. Standardized bioactivity assays with quantitative EC50 determination should be performed on each preparation to enable meaningful comparisons. The His-tag's potential influence on protein function necessitates parallel experiments with both tagged and enzymatically de-tagged versions to distinguish genuine CXCL1 effects from artifacts. Environmental factors significantly impact CXCL1 stability and activity; therefore, researchers should standardize buffer compositions, pH, temperature, and experimental timing. Adsorption to laboratory plasticware poses another challenge, which can be addressed by including carrier proteins (0.1% BSA) in working solutions and using low-protein-binding materials. For cell-based assays, passage number and culture conditions of target cells should be carefully controlled, as receptor expression levels can vary with cell state. Detailed methodological reporting in publications is essential, including specific catalog numbers of commercial reagents, exact buffer compositions, and comprehensive validation data. Collaborative standardization efforts across laboratories, including round-robin testing and development of reference standards, would substantially advance reproducibility in CXCL1 research.
Detecting and quantifying native CXCL1 in biological samples presents substantial analytical challenges requiring specialized approaches. The protein's low physiological concentration (typically pg/mL to ng/mL range in plasma) necessitates highly sensitive detection methods, with ELISA remaining the gold standard despite limitations in distinguishing between active and inactive forms. Matrix effects from complex biological samples can significantly interfere with accurate quantification, requiring optimized sample preparation protocols that may include immunoprecipitation steps prior to analysis. CXCL1's binding to extracellular matrix components and cell surface proteoglycans means that soluble measurements may not reflect the total biologically relevant pool, necessitating extraction protocols that release matrix-bound chemokines. Post-translational modifications, particularly N-terminal processing by proteases like matrix metalloproteinases, dramatically alter CXCL1 activity but are difficult to distinguish with most antibody-based detection methods. Researchers addressing these challenges should consider implementing mass spectrometry-based approaches that can identify specific CXCL1 isoforms and modifications, proximity ligation assays to detect CXCL1 in tissue contexts while preserving spatial information, and aptamer-based detection systems that offer improved sensitivity over traditional antibody methods. For functional quantification, bioactivity assays measuring neutrophil chemotaxis or calcium flux should complement immunological detection methods. Standardization of collection protocols is critical, as CXCL1 levels can be artificially elevated by improper sample handling that activates platelets or neutrophils, which release stored CXCL1 upon stimulation.
Resolving data contradictions in CXCL1 research requires systematic methodological approaches that address the multifaceted nature of chemokine biology. The observed phenomenon in which CXCL1 stimulates cancer cell growth in vitro but inhibits tumor growth in vivo (similar to findings reported for MR409 in search result ) exemplifies such contradictions that demand careful investigation. To resolve such discrepancies, researchers should implement parallel in vitro and in vivo models using identical CXCL1 preparations to directly compare outcomes. Concentration-dependent effects require particular attention, as CXCL1 often exhibits bell-shaped dose-response curves where both too little and too much activity yield similar phenotypic outcomes through different mechanisms. Time-course experiments are essential, as immediate CXCL1 effects may differ substantially from long-term consequences. Cell-type specific responses should be systematically evaluated, as the same CXCL1 signal may elicit opposing effects in different cell populations within a tissue. Context-dependency, particularly regarding the presence of other inflammatory mediators or matrix components, should be methodically varied to identify conditional effects. For contradictions between human and animal studies, humanized mouse models or human tissue explants can provide valuable intermediate systems. When contradictions persist despite methodological optimization, computational modeling integrating multiple datasets can help identify parameter spaces where seemingly contradictory observations become reconcilable. Meta-analysis approaches comparing methodological details across contradictory studies often reveal critical procedural differences that explain divergent outcomes. Finally, comprehensive reporting of negative results alongside positive findings is essential for building an accurate understanding of CXCL1 biology.
Several emerging technologies show promise for significantly advancing CXCL1 research in the coming decade. CRISPR-based approaches beyond simple knockout models, including base editing and epigenetic modifiers, will enable precise manipulation of CXCL1 expression and regulatory elements in physiologically relevant contexts. Single-cell multi-omics technologies will reveal cell-specific responses to CXCL1 stimulation across transcriptome, proteome, and metabolome levels, providing unprecedented insight into signaling heterogeneity. Advanced imaging technologies, particularly light sheet microscopy combined with chemokine-sensitive biosensors, will allow real-time visualization of CXCL1 gradients and cellular responses in three-dimensional tissues. Organ-on-chip platforms incorporating microfluidic gradient generators will enable controlled studies of CXCL1-directed cell migration in physiologically relevant microenvironments. In structural biology, cryo-electron microscopy advances will likely reveal CXCL1-receptor complexes at atomic resolution, informing structure-based drug design. Artificial intelligence approaches, particularly deep learning applied to multi-dimensional datasets, will help identify previously unrecognized patterns in CXCL1 signaling networks. For translational applications, mRNA and lipid nanoparticle technologies may enable targeted delivery of modified CXCL1 or antagonists to specific tissues. Researchers should strategically incorporate these technologies while maintaining focus on fundamental biological questions, as technological sophistication alone cannot substitute for well-designed experiments addressing clearly defined hypotheses about CXCL1 function.
Interdisciplinary approaches offer tremendous potential to enhance our understanding of CXCL1 biology by addressing its multifaceted functions from diverse perspectives. Collaborations between immunologists and neuroscientists could further elucidate CXCL1's role in neuroimmune interactions, particularly relevant given its documented functions in both immune cell recruitment and oligodendrocyte precursor migration . Integrating systems biology with traditional biochemistry would enable modeling of complex CXCL1 signaling networks while maintaining mechanistic rigor regarding specific molecular interactions. The incorporation of biophysical approaches, particularly techniques measuring protein diffusion and gradient formation, would provide critical insights into how CXCL1 spatial distribution influences its biological effects. Computational biology collaborations could develop predictive models of CXCL1 activity across different tissues and disease states, generating testable hypotheses for experimental validation. Partnerships with clinical researchers would enhance translational relevance by connecting basic CXCL1 biology to human disease mechanisms through analysis of patient samples. Material science collaborations could develop advanced delivery systems for CXCL1 modulators or biomaterials incorporating CXCL1 for tissue engineering applications. Creating interdisciplinary research teams requires institutional support for collaborative projects, shared technical resources, and training opportunities that familiarize researchers with methodologies outside their primary discipline. The most successful interdisciplinary approaches will maintain a clear focus on specific CXCL1-related questions while leveraging diverse expertise to address these questions from multiple angles simultaneously.
CXCL1-based therapeutic approaches in regenerative medicine show significant potential across multiple tissue contexts. In wound healing applications, controlled delivery of CXCL1 could optimize neutrophil recruitment during the inflammatory phase while preventing excessive neutrophil accumulation that delays resolution. For cardiovascular regeneration, CXCL1's angiogenic properties could be harnessed to improve vascularization of engineered tissues or to promote revascularization following ischemic injury. In neural tissue regeneration, CXCL1's influence on oligodendrocyte precursor migration suggests applications in promoting remyelination, though careful timing and dosing would be essential given its reported inhibitory effects on these cells in some contexts . For musculoskeletal regeneration, emerging evidence suggesting CXCL1 roles in mesenchymal stem cell recruitment and differentiation presents opportunities for enhancing fracture healing or cartilage repair. Development of these therapeutic approaches requires addressing several technical challenges, including: creating delivery systems that achieve physiologically relevant CXCL1 gradients rather than bolus dosing; developing modified CXCL1 variants with enhanced stability or altered receptor specificity for specific regenerative applications; and designing combinatorial approaches that integrate CXCL1 with other growth factors in temporally controlled sequences matching natural healing processes. Preclinical testing should include both acute injury models and chronic disease models to determine context-specific efficacy, with particular attention to potential pro-inflammatory side effects. Successful translation will require close collaboration between protein engineers, biomaterial scientists, and clinicians specializing in specific regenerative applications.
Property | GRO-Alpha/CXCL1 | GRO-Beta/CXCL2 | GRO-Gamma/CXCL3 | IL-8/CXCL8 |
---|---|---|---|---|
Molecular Weight | 8-11 kDa | 7-8 kDa | 7-8 kDa | 8-9 kDa |
Primary Receptor | CXCR2 (high affinity), CXCR1 (low affinity) | CXCR2 | CXCR2 | CXCR1, CXCR2 |
ELR Motif | Present | Present | Present | Present |
Primary Cellular Sources | Melanoma cells, macrophages, neutrophils, epithelial cells | Macrophages, epithelial cells | Macrophages, fibroblasts | Macrophages, epithelial cells, fibroblasts |
Neutrophil Chemotaxis Potency | High | High | Moderate | Very High |
Role in Angiogenesis | Pro-angiogenic | Pro-angiogenic | Pro-angiogenic | Pro-angiogenic |
Known Roles in Development | Inhibits oligodendrocyte precursor migration | Limited evidence | Limited evidence | Limited evidence |
GAG Binding Affinity | Moderate | Low | Low | High |
Dimerization Tendency | Forms dimers at high concentrations | Forms dimers at high concentrations | Forms dimers at high concentrations | Forms dimers at physiological concentrations |
This table synthesizes information from multiple sources to provide comparative information on GRO-Alpha/CXCL1 and related chemokines, highlighting their structural and functional similarities and differences .
Research Area | Experimental Model | CXCL1 Concentration Range | Key Findings | Methodological Considerations |
---|---|---|---|---|
Cancer Biology | Human melanoma cell lines | 1-100 ng/mL | Promotes proliferation and invasion in vitro | Serum-free conditions required; matrix components alter response |
Inflammation | Mouse models of acute lung injury | 0.1-1 μg/animal (intranasal) | Neutrophil recruitment; increased vascular permeability | Timing of administration critical; effects dose-dependent |
Neurodevelopment | Primary oligodendrocyte precursor cultures | 10-500 ng/mL | Inhibits migration; affects differentiation timing | Growth factor context influences outcomes |
Angiogenesis | Endothelial tube formation assay | 5-50 ng/mL | Promotes tubule formation; synergizes with VEGF | Requires presence of supporting matrix |
Wound Healing | Excisional wound models | 0.1-1 μg/wound (topical) | Initial acceleration followed by potential delayed resolution | Administration schedule affects outcome |
Protein Structure Studies | NMR spectroscopy | 0.5-2 mg/mL (^15N/^13C labeled) | Dimerization at high concentrations affects receptor binding | pH and salt concentration affect structural states |
This table compiles experimental applications of recombinant CXCL1 across different research models, providing practical guidance on concentration ranges and methodological considerations for researchers designing experiments .
Artifact | Underlying Cause | Detection Method | Recommended Solution |
---|---|---|---|
False negative in bioactivity assays | Protein adsorption to plasticware | Quantify protein before and after incubation in experimental vessels | Include carrier protein (0.1% BSA); use low-binding plasticware |
Inconsistent chemotaxis results | Variable receptor expression on target cells | Flow cytometry for CXCR2 levels | Standardize cell culture conditions; sort cells for consistent receptor expression |
Apparent loss of activity during storage | Oxidation of critical cysteine residues | Mass spectrometry to detect oxidized forms | Include reducing agents in storage buffer; aliquot and store under nitrogen |
Enhanced activity of His-tagged vs. native protein | Altered protein-protein interactions or receptor binding | Direct comparison experiments with tagged and untagged protein | Use enzymatic tag removal for critical experiments; validate with both forms |
Contradictory in vitro vs. in vivo results | Absence of GAG interactions in simplified in vitro systems | Compare standard assays with GAG-binding mutants | Include relevant GAG components in in vitro systems |
Batch-to-batch variation | Post-translational modification differences | Isoelectric focusing; reverse phase HPLC | Implement standardized production and purification protocols; pool multiple small batches |
This table identifies common methodological artifacts encountered in CXCL1 research, their underlying causes, and recommended solutions to enhance experimental reliability and reproducibility. The information is synthesized from research experience and best practices in the chemokine field .
GRO-Alpha, also known as Growth-Regulated Oncogene Alpha (GRO-Alpha) or Melanoma Growth Stimulatory Activity Alpha (MGSA), is a chemokine belonging to the CXC chemokine family. It is encoded by the CXCL1 gene and plays a crucial role in various physiological and pathological processes, including inflammation, wound healing, and cancer progression.
GRO-Alpha is a small cytokine with a molecular weight of approximately 7.8 kDa . It consists of 73 amino acids and includes the ‘ELR’ motif, which is characteristic of the CXC chemokine family . The recombinant form of GRO-Alpha, often tagged with a His (histidine) tag for purification purposes, is produced in Escherichia coli (E. coli) expression systems .
GRO-Alpha is primarily known for its role in the recruitment and activation of neutrophils, a type of white blood cell essential for the immune response . It binds with high affinity to the IL-8 receptor type B (CXCR2), leading to the activation and chemotaxis of neutrophils to sites of tissue injury or infection . This chemokine is also involved in angiogenesis, the formation of new blood vessels, which is critical for wound healing and tumor growth .
GRO-Alpha has been implicated in various diseases, including cancer, inflammatory conditions, and autoimmune disorders. Its overexpression has been observed in several types of cancer, such as melanoma, where it promotes tumor growth and metastasis . Additionally, GRO-Alpha is involved in the pathogenesis of inflammatory diseases like rheumatoid arthritis and chronic obstructive pulmonary disease (COPD) .
Recombinant GRO-Alpha is widely used in research to study its biological functions and therapeutic potential. It is utilized in various assays, including chemotaxis assays, to investigate its role in immune cell recruitment and activation . The His tag facilitates the purification and detection of the recombinant protein, making it a valuable tool for biochemical and structural studies .