SCRN1 Antibody targets the secernin 1 protein, a 46 kDa molecular weight protein encoded by the SCRN1 gene (Gene ID: 9805). This antibody is widely used in research to study SCRN1's role in secretory pathways and its implications in diseases such as cancer and autoimmune disorders .
Western Blot (WB): Detects SCRN1 at ~50 kDa in human A549 and HeLa cells, as well as mouse brain tissue .
Immunohistochemistry (IHC): Validated in human stomach cancer tissue with recommended antigen retrieval using TE buffer (pH 9.0) .
Immunofluorescence (IF) and ELISA: Utilized for subcellular localization and quantitative assays .
SCRN1 is part of the PM/Scl complex, which is targeted by autoantibodies in systemic sclerosis (SSc) and overlap syndromes like polymyositis-scleroderma .
Anti-PM/Scl antibodies are associated with:
SCRN1 is overexpressed in gastric and colorectal cancers, where it regulates secretory pathways critical for tumor progression .
| Feature | Proteintech (14303-1-AP) | EMD Millipore (ABC829) |
|---|---|---|
| Host | Rabbit IgG | Rabbit IgG |
| Applications | WB, IHC, CoIP, ELISA | WB, IHC, IF |
| Tissue Validation | Human stomach cancer, mouse brain | Broad tissue specificity |
| Cited Reactivity | Human, Mouse, Rat | Human, Mouse |
KEGG: ath:AT4G10457
UniGene: At.63294
SCRL1 Antibody represents a specific class of antibodies that recognize and bind to target antigens, similar to how antibodies function generally in the immune system. Fundamentally, antibodies are proteins produced by the immune system that protect against disease by recognizing specific foreign substances called pathogens, such as viruses and bacteria. In the context of research, antibodies like SCRL1 can be utilized to detect, isolate, or manipulate specific biological molecules of interest. SCRL1 Antibody works by recognizing specific epitopes (binding sites) on target antigens, creating an antibody-antigen complex that can be detected or used in various experimental applications. This recognition property makes antibodies invaluable tools in numerous research techniques including western blotting, immunohistochemistry, flow cytometry, and ELISA .
SCRL1 Antibody serology tests, like other antibody serology tests, are designed to detect the presence of specific antibodies in blood samples rather than directly detecting pathogens or antigens. They differ from molecular diagnostic methods such as PCR, which detect the genetic material of pathogens. SCRL1 Antibody serology assays typically utilize enzyme-linked immunosorbent assay (ELISA), chemiluminescent immunoassay, or other immunoassay techniques to detect antibodies. Unlike direct pathogen detection methods, antibody serology tests indicate whether a person has been previously exposed to or vaccinated against a particular pathogen rather than diagnosing active infection. This makes them particularly useful for epidemiological studies, monitoring immune status, or assessing vaccination efficacy .
For research applications of SCRL1 Antibody testing, blood samples are most commonly used, specifically serum or plasma. The collection process typically involves a healthcare professional taking a blood sample from a vein in the subject's arm using a small needle. After collection, the blood sample is processed to separate the serum or plasma from cellular components. In specialized research applications, SCRL1 Antibody testing may also be performed on other biological fluids such as cerebrospinal fluid, bronchoalveolar lavage fluid, or tissue homogenates depending on the specific research question. The selection of sample type should be guided by the research objectives, the expected concentration of the target antibody, and the biological compartment most relevant to the research question .
Validating the specificity of SCRL1 Antibody requires a systematic approach incorporating multiple experimental techniques. Begin with western blot analysis using cell or tissue lysates expressing and not expressing the target protein to confirm that the antibody binds to a protein of the expected molecular weight. Perform immunoprecipitation followed by mass spectrometry to identify all proteins bound by the antibody. Include knockout or knockdown controls in your experimental system—the antibody signal should be absent or significantly reduced in samples lacking the target protein. Use competitive binding assays with purified antigen to demonstrate specific displacement of antibody binding. For advanced validation, implement peptide array analysis to map the exact epitope recognized by the antibody. Cross-reactivity testing with structurally similar proteins will further confirm specificity. Document all validation steps methodically, as antibody specificity is crucial for accurate interpretation of experimental results .
Isolating and characterizing novel SCRL1 Antibody variants requires a sophisticated multi-step approach. Begin with peripheral blood mononuclear cell (PBMC) isolation from subjects of interest using density gradient centrifugation. Implement fluorescence-activated cell sorting (FACS) to isolate B cells expressing SCRL1 Antibody by utilizing labeled antigens as probes—specifically target CD19+/CD20+/IgM–/IgA+ or IgG+ B cells that bind to your antigen of interest. Single-cell RNA sequencing of sorted B cells will identify transcripts encoding antibody heavy and light chains. Recover paired heavy- and light-chain antibody sequences, which typically yields approximately 80% success rate based on comparable antibody isolation projects. Express selected antibodies recombinantly and screen using sensitive binding assays such as MSD (Meso Scale Discovery) to assess binding to relevant antigens and epitopes. Characterize binding kinetics using surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to determine association and dissociation rates and calculate affinity constants. Functional characterization should include neutralization assays using both pseudovirus and live virus systems when applicable .
Integrating SCRL1 Antibody into multiplex panels requires strategic panel design that considers marker expression levels, spectral overlap, and biological relevance. Start by using single-cell RNA-sequencing data to identify optimal marker combinations that can distinguish cell populations of interest. Utilize interactive platforms like Cytomarker that enable human-in-the-loop design of antibody panels from transcriptomic data. When incorporating SCRL1 Antibody, assess potential antigen density on target cells to determine appropriate fluorophore brightness needed. Validate the panel through systematic testing of SCRL1 Antibody alongside other markers using methods such as Cytometry by Time-of-Flight (CyTOF) or spectral flow cytometry. Consider developing a novel antibody screening strategy similar to approaches used in recent studies where arrayed cell populations are labeled with unique cell surface barcodes and experimental markers of interest. This allows simultaneous validation of multiple markers across different cell populations. Perform correlation analysis between RNA expression and antibody signal to validate the specificity and performance of SCRL1 Antibody in your panel. Assess positive predictive value, sensitivity, and specificity of SCRL1 Antibody detection in comparison to transcriptomic data .
The binding affinity and specificity of SCRL1 Antibody are determined by precise structural characteristics of both the antibody and its target epitope. The complementarity-determining regions (CDRs), particularly within the heavy chain variable domain, form the primary antigen-binding pocket. X-ray crystallography and cryo-electron microscopy studies of antibody-antigen complexes reveal that high-affinity binding often involves multiple non-covalent interactions including hydrogen bonds, van der Waals forces, salt bridges, and hydrophobic interactions. The three-dimensional arrangement of these interaction sites creates a binding interface with high geometric and chemical complementarity to the target epitope. Germline gene origin significantly influences binding properties—for instance, antibodies derived from IGHV1-58 germline have demonstrated exceptional neutralization potency in some systems. Somatic hypermutation introduces critical modifications that fine-tune binding affinity, with as few as 1-3 amino acid substitutions potentially increasing affinity by several orders of magnitude. For optimal binding, SCRL1 Antibody likely recognizes conformational epitopes that minimize contact with mutational hotspots, enhancing cross-reactivity to variant forms of the target. This structural understanding is essential for engineering antibodies with improved binding characteristics .
Potential escape mechanisms against SCRL1 Antibody include mutations within the target epitope, allosteric conformational changes in the antigen, masking of epitopes by post-translational modifications, and altered expression of the target protein. To identify these mechanisms experimentally, apply antibody selection pressure to replication-competent model systems expressing the target antigen, similar to methods used with vesicular stomatitis virus (VSV) expressing viral antigens. Using next-generation sequencing, monitor emerging mutations that confer resistance to SCRL1 Antibody. Structural analysis of these escape mutants by cryo-electron microscopy can reveal precise mechanisms of resistance. To prevent escape, implement a combinatorial approach using multiple antibodies targeting non-overlapping epitopes on the same antigen. Design antibody combinations with complementary modes of recognition based on competition assays and structural data. Target conserved regions with functional constraints, where mutations would compromise the antigen's essential functions. Engineer bispecific or multispecific antibodies that simultaneously engage multiple epitopes, significantly reducing the probability of escape. For therapeutic applications, consider antibody cocktails that recognize different domains of the target, as this strategy has demonstrated effective prevention of escape in viral systems .
Designing robust cross-reactivity experiments for SCRL1 Antibody requires a comprehensive, multi-platform approach. Begin with computational analysis by aligning the target antigen sequence with potential cross-reactive proteins to identify regions of homology. Develop a protein microarray containing the target antigen, closely related proteins, and structurally similar but functionally distinct proteins to assess binding specificity across a broad spectrum. Implement surface plasmon resonance (SPR) to quantitatively measure binding kinetics to both target and potential cross-reactive antigens, calculating affinity constants for each interaction. Perform immunoprecipitation followed by mass spectrometry (IP-MS) using lysates from relevant biological systems to identify all proteins captured by SCRL1 Antibody. For cellular systems, conduct flow cytometry with cells expressing the target antigen and cells expressing related proteins, comparing binding profiles. Deploy competition assays where unlabeled potential cross-reactive antigens are used to compete with labeled target antigen for antibody binding. Include appropriate controls such as isotype-matched control antibodies and pre-absorption studies. Document all cross-reactivity in detail, as even minor cross-reactivity can significantly impact experimental interpretation and therapeutic applications .
Developing a high-throughput screening system for SCRL1 Antibody candidates requires integration of multiple technologies. Implement a hybridoma-based or phage display approach to generate a diverse library of potential antibody candidates. Design a primary screening assay using ELISA or bead-based multiplex systems to rapidly assess binding to the target antigen across thousands of candidates. Develop a secondary functional screen that evaluates candidates for specific desired activities such as neutralization, receptor blocking, or internalization. Incorporate automation through robotic liquid handling systems and plate readers with data management software for rapid processing and analysis. Design the screening cascade to progressively narrow candidates through increasingly stringent criteria, including cross-reactivity testing, epitope binning, and affinity determination. Implement machine learning algorithms to identify patterns in antibody sequence and function relationships from screening data, enhancing subsequent screening rounds. Include relevant physiological conditions in later screening stages to ensure antibody performance in the intended research environment. For higher confidence in results, develop a novel antibody screening strategy similar to recent approaches where >200 candidates can be simultaneously evaluated against multiple cell types using unique cell surface barcodes and selective labeling with experimental markers .
To preserve SCRL1 Antibody reactivity in tissue samples for immunohistochemistry, implement a comprehensive protocol addressing each critical phase. Begin with proper fixation—use 10% neutral buffered formalin for 24-48 hours at room temperature, avoiding over-fixation which can mask epitopes. For antigen retrieval, optimize conditions by testing multiple methods: heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0), EDTA buffer (pH 9.0), or enzymatic retrieval with proteinase K. Perform systematic blocking using 3% BSA with 5% horse serum in TBS for 60 minutes at room temperature to minimize non-specific binding. Antibody concentration must be empirically determined; start with 1:100 dilution and titrate as needed. Incubate primary antibody overnight at 4°C in blocking solution to maximize specific binding while minimizing background. For detection, compare direct detection with conjugated antibodies versus multi-step approaches using secondary antibodies. If using multiplexed staining, implement proper controls for each antibody combination and perform sequential staining with intermediate washing steps. For validation, include positive and negative tissue controls, isotype controls, and absorption controls using the immunizing peptide. Document all optimization steps meticulously for reproducibility across experiments and facilities .
Accurate quantification of SCRL1 Antibody binding kinetics requires sophisticated biophysical techniques and careful experimental design. Surface plasmon resonance (SPR) serves as the gold standard method—immobilize the target antigen on a sensor chip using amine coupling chemistry or capture approaches that preserve native conformation. Prepare a concentration series of SCRL1 Antibody spanning at least two orders of magnitude around the expected KD value. Record association and dissociation phases at multiple antibody concentrations and fit the resulting sensorgrams to appropriate binding models (1:1 Langmuir binding, heterogeneous ligand, etc.). Calculate association rate constant (ka), dissociation rate constant (kd), and equilibrium dissociation constant (KD = kd/ka). Bio-layer interferometry (BLI) offers an alternative label-free approach with similar principles but different detection technology. For solution-based measurements, implement isothermal titration calorimetry (ITC) or microscale thermophoresis (MST). Validate results across multiple techniques and experimental conditions to ensure robustness. Assess the impact of pH, ionic strength, and temperature on binding parameters to understand the physiological relevance of the interaction. For antibody fragments (Fabs), comparison with full antibody binding can provide insights into avidity effects. Reported affinities for high-quality research antibodies typically fall in the nanomolar range (2.3 to 7.3 nM), with exceptional antibodies achieving sub-nanomolar affinities .
Antibody research has significant applications in understanding autoimmune disease mechanisms, with methodologies applicable to studies involving SCRL1 Antibody. In autoimmune conditions, antibodies raised against self-antigens play crucial roles in pathogenesis. Research approaches have demonstrated how anti-peptide antibodies can be raised to recognize neo-antigens created during protein degradation or modification. For example, studies of erythrocyte complement receptor 1 (CR1) in systemic lupus erythematosus (SLE) utilized antibodies raised against peptide sequences corresponding to regions between protein domains. These antibodies successfully recognized proteolysed CR1 remnants on erythrocytes, providing insights into the mechanisms of CR1 level reduction in diseases with in vivo complement activation. Similar methodological approaches could be applied with SCRL1 Antibody to investigate neo-antigens formed during autoimmune processes. The technique involves raising antibodies to peptides corresponding to specific protein sequences that become exposed following proteolysis or other modifications. Such antibodies serve as valuable diagnostic tools for detecting modified proteins in clinical samples, though their development requires optimization to achieve sufficient affinity for reliable detection. This approach enables investigation of protein modification mechanisms in autoimmune pathology, potentially revealing new therapeutic targets .
Engineering antibodies for enhanced specificity involves several sophisticated approaches applicable to SCRL1 Antibody research. Begin with comprehensive epitope mapping using hydrogen-deuterium exchange mass spectrometry or peptide array analysis to precisely identify the binding site. Implement site-directed mutagenesis of complementarity-determining regions (CDRs), particularly in the heavy chain, to modify amino acids involved in non-specific interactions while preserving target binding. Utilize computational structure-based design to predict mutations that would enhance shape complementarity to the target epitope. Apply directed evolution techniques such as phage display with stringent selection conditions—including multiple rounds of negative selection against similar proteins—to eliminate cross-reactive variants. Consider CDR grafting, where the binding regions from SCRL1 Antibody are transferred to a different antibody framework with superior stability characteristics. For research applications requiring extreme specificity, develop bispecific formats that require simultaneous binding to two distinct epitopes on the target protein. Implement affinity maturation through targeted diversification of CDR residues followed by screening under increasingly stringent conditions. Systematically evaluate engineered variants using multiple binding assays, cross-reactivity panels, and functional tests to ensure that specificity enhancements don't compromise essential functions. Document the engineering process comprehensively, as understanding the molecular basis for specificity improvements contributes valuable insights to antibody engineering science .
When evaluating antibody performance, comprehensive comparative analysis across multiple parameters is essential. While specific comparative data for SCRL1 Antibody would require detailed experimentation, the methodological approach involves assessing binding affinity, specificity, and functional activity against benchmark antibodies. High-performing research antibodies typically demonstrate nanomolar affinity to their targets, with elite antibodies achieving KD values in the 2-7 nM range as measured by surface plasmon resonance or bio-layer interferometry. Neutralization potency represents another critical performance metric, with IC50 values for potent neutralizing antibodies typically ranging from 2-70 ng/ml in pseudovirus assays and 2-5 ng/ml in live virus neutralization systems. The capacity to recognize diverse variant forms of the target is increasingly important, particularly for targets that demonstrate natural variation or mutation. Antibodies derived from specific germline genes, such as IGHV1-58, have demonstrated superior cross-variant recognition in some systems. Structural studies of antibody-antigen complexes provide insights into binding modes that confer broad recognition capabilities. For research applications, comparison should include performance across different experimental techniques (western blot, immunoprecipitation, flow cytometry, IHC) as certain antibodies excel in specific applications due to their recognition of particular conformational states of the target protein .
Emerging technologies poised to revolutionize antibody characterization include advanced structural biology approaches, high-throughput functional screening, and computational prediction methods. Cryo-electron microscopy advances now enable visualization of antibody-antigen complexes at near-atomic resolution without crystallization, providing unprecedented structural insights into binding mechanisms. Single-cell technologies are evolving to simultaneously analyze transcriptomics, proteomics, and functional properties of individual B cells, enabling direct correlation between antibody sequence and function. High-throughput antibody screening approaches using cell surface barcoding and mass cytometry allow simultaneous evaluation of hundreds of antibodies against diverse cell populations, dramatically accelerating discovery and characterization workflows. Computational antibody design is advancing through machine learning algorithms trained on antibody-antigen interaction databases, enabling in silico prediction of binding properties and optimization of antibody sequences. Automated antibody discovery platforms integrating microfluidics, single-cell isolation, sequencing, and expression systems are streamlining the pipeline from B cell isolation to characterized antibody. For spatial applications, multiplexed imaging techniques using iterative antibody staining or mass cytometry imaging permit visualization of dozens of targets simultaneously in tissue contexts. These technological advances collectively promise to enhance the depth, speed, and precision of antibody characterization while reducing resource requirements .
Antibody research provides crucial insights for universal vaccine development against mutable pathogens, with broadly applicable principles. The discovery of broadly neutralizing antibodies (bNAbs) that recognize conserved epitopes across variant forms of pathogens represents a key strategy. For example, recent research identified antibody SC27, which neutralizes all known variants of SARS-CoV-2 by recognizing highly conserved regions of the spike protein. Similar approaches could be applied to identify broadly neutralizing antibodies against other variable pathogens. Molecular characterization of such antibodies reveals critical binding sites that remain conserved despite mutation, providing valuable templates for vaccine design. Structural analysis using cryo-electron microscopy can precisely map these conserved epitopes, enabling the design of immunogens that specifically present these regions to the immune system. By studying the germline origins of broadly neutralizing antibodies, researchers can develop vaccination strategies that specifically activate B cell lineages with the potential to develop broad protection. The goal of such vaccinology approaches is to work toward universal vaccines that generate antibodies with broad protection against rapidly mutating viruses, rather than strain-specific responses. This research direction represents one of the most promising frontiers in vaccine development, with potential applications against diverse pathogens including influenza, HIV, and coronaviruses .