LRPAP1 antibodies bind to LRPAP1, a 39 kDa protein that facilitates the proper folding and trafficking of LDL receptor family members (e.g., LRP1) from the endoplasmic reticulum to the plasma membrane . By inhibiting LRPAP1’s interaction with these receptors, antibodies can modulate receptor-mediated processes such as:
Phagocytosis (e.g., microglial clearance of amyloid-beta (Aβ) in Alzheimer’s disease)
Autoantibody-driven signaling (e.g., in mantle cell lymphoma)
LRPAP1 antibodies are used to study microglial dysfunction in Alzheimer’s disease. Stressed microglia release LRPAP1, which inhibits phagocytosis of Aβ plaques and synapses at concentrations as low as 10 nM . Key findings include:
Aβ aggregation: LRPAP1 reduces Aβ fibrillization in vitro at 500 nM (10:1 molar ratio to Aβ) .
Therapeutic potential: Neutralizing LRPAP1 antibodies could enhance microglial clearance of Aβ, though physiological relevance requires further validation .
LRPAP1 autoantibodies are biomarkers in MCL, associated with improved patient outcomes:
Mechanism: These autoantibodies are clonally restricted (κ or λ light chains) and correlate with reduced hazard ratios for disease progression (HR = 0.48 for FFS; HR = 0.47 for OS) .
LRPAP1 antibodies counteract viral immune evasion strategies. For example:
Enterovirus 71 (EV71): Secreted LRPAP1 binds IFNAR1, triggering its degradation and suppressing antiviral responses. Anti-LRPAP1 antibodies restore IFNAR1 levels and reduce viral load .
The LRPAP1 Rabbit Polyclonal Antibody (CAB13026) is widely used to detect LRPAP1 in Western blotting. Key features include:
Specificity: Targets human LRPAP1 with minimal cross-reactivity .
Applications: Studying cholesterol metabolism, receptor trafficking, and pathological aggregates in neurodegeneration .
LRPAP1 antibodies exert effects through multiple pathways:
Receptor internalization: Anti-LRPAP1 antibodies induce rapid lysosomal degradation of LRP1 in microglia, impairing phagocytosis .
Ligand competition: LRPAP1 blocks ligand binding to LDL receptor domains II, III, and IV, inhibiting signaling cascades .
Immune modulation: In MCL, autoantibodies may paradoxically enhance survival by altering B-cell receptor signaling .
Alzheimer’s disease: Antibodies targeting LRPAP1 could enhance Aβ clearance but risk exacerbating neuroinflammation .
Oncology: LRPAP1 autoantibodies may serve as prognostic markers or immunotherapeutic targets in MCL .
Antiviral strategies: Neutralizing extracellular LRPAP1 could restore interferon signaling during viral infections .
Dose dependency: Effects of LRPAP1 antibodies vary with concentration (e.g., 50–200 nM rLRPAP1 reduces IFNAR1 within 1 hour) .
Physiological relevance: Most studies use supraphysiological LRPAP1 levels; in vivo validation is critical .
Dual roles: LRPAP1 antibodies may have opposing effects in different diseases, necessitating context-specific evaluation .
LRPAP1 (Low density lipoprotein receptor-related protein associated protein 1) is a 39 kDa glycoprotein that functions as a chaperone protein encoded by the LRPAP1 gene in humans. The significance of LRPAP1 lies in its crucial role in the trafficking of certain members of the LDL receptor family, particularly LRP1 and LRP2. This protein binds to the alpha-2-macroglobulin receptor and other members of the low density lipoprotein receptor family, acting to inhibit the binding of all known ligands for these receptors. LRPAP1's molecular chaperone function prevents receptor aggregation and degradation in the endoplasmic reticulum, making it an important target for studies focused on lipoprotein metabolism, receptor trafficking, and associated pathologies . Recent research has also identified LRPAP1 as a B-cell receptor autoantigen in certain lymphomas, expanding its significance to cancer research and potential therapeutic applications .
When conducting Western Blot assays with LRPAP1 antibodies, researchers should follow a methodical approach to achieve optimal results. First, proteins should be separated on a 10% SDS-PAGE gel and transferred to a PVDF membrane using a semi-dry transfer system. After transferring, block the membrane overnight at 4°C in PBS with 10% nonfat dry milk to reduce non-specific binding. For detection using commercially available rabbit polyclonal antibodies to LRPAP1, incubate the membrane with the primary antibody at an appropriate dilution (typically 1:2000) for 1 hour at room temperature . Follow with an HRP-labeled secondary antibody (anti-rabbit IgG) at a 1:3000 dilution. For visualization, use a chemiluminescence reagent appropriate for immunoblot detection. When working with tagged LRPAP1 constructs, detection can be achieved using tag-specific antibodies, such as anti-His antibodies (for His-tagged proteins) or ANTI-FLAG M2 antibodies (for FLAG-tagged proteins) . Store antibodies at -20°C or -80°C and avoid repeated freeze-thaw cycles to maintain antibody integrity and performance .
LRPAP1 antibodies have been validated for multiple experimental applications including Enzyme-Linked Immunosorbent Assay (ELISA), Immunohistochemistry (IHC), and Western Blotting (WB) . When designing experiments, appropriate positive controls should be selected based on known LRPAP1 expression patterns. For human samples, cell lines with confirmed LRPAP1 expression such as MAVER1 and Z138 mantle cell lymphoma cells can serve as positive controls, particularly for studies related to B-cell receptor interactions . For studies in rodent models, the antibody reactivity extends to mouse and rat samples, as commercial LRPAP1 antibodies show cross-reactivity across these species . When designing immunohistochemistry experiments, tissues with endogenous LRPAP1 expression, such as liver tissues where LDL receptor family members are abundant, should be considered as positive controls. For negative controls, cell lines known not to express LRPAP1-reactive B-cell receptors, such as Granta-519 and Mino cell lines, can be utilized to confirm binding specificity . Always validate antibody performance in your specific experimental system before proceeding with definitive experiments.
To maintain optimal activity of LRPAP1 antibodies, proper storage and handling procedures are critical. Upon receipt, store antibodies at -20°C or -80°C in accordance with manufacturer recommendations . Avoid repeated freeze-thaw cycles as this can significantly degrade antibody quality and performance. When working with the antibody, aliquot stock solutions into smaller volumes to minimize freeze-thaw events. The storage buffer composition is important for stability – typical formulations include pH 7.4 PBS with 0.05% sodium azide (NaN3) and 40% glycerol . The glycerol acts as a cryoprotectant, while sodium azide prevents microbial contamination during storage. When handling antibodies, use sterile techniques and maintain cold chain conditions whenever possible. Prior to use, centrifuge the antibody vial briefly to collect the solution at the bottom of the tube. Dilute antibodies in appropriate buffers immediately before use and do not store diluted antibodies for extended periods. For long-term stability assessment, periodically test antibody activity using standardized assays to ensure performance has not deteriorated during storage.
LRPAP1 antibodies can be strategically employed to investigate B-cell receptor (BCR) autoantigen interactions in mantle cell lymphoma (MCL) through multiple sophisticated approaches. Begin by using flow cytometry to identify MCL cell lines with LRPAP1-reactive BCRs – incubate 5 × 10^6 MCL cells (such as MAVER1 or Z138) with 5-10 μg/mL of LRPAP1 antibodies or LRPAP1-containing constructs at 4°C for 30 minutes, followed by appropriate fluorescently-labeled secondary antibodies . Compare binding patterns with non-LRPAP1-reactive MCL cell lines (like Granta-519 and Mino) to establish specificity. For mechanistic studies, epitope mapping experiments using truncated LRPAP1 constructs can identify the precise reactive regions – research has shown that amino acids 264-318 of LRPAP1 contain the critical epitope recognized by MCL BCRs . To study functional consequences of LRPAP1-BCR interactions, design competition assays where purified LRPAP1 protein or peptides compete with anti-LRPAP1 antibodies for BCR binding, providing insights into binding kinetics and affinities. Immunoprecipitation experiments using anti-LRPAP1 antibodies can isolate BCR complexes from MCL cells, enabling proteomic analysis of associated signaling components. Ultimately, these approaches allow researchers to characterize the molecular basis of autoantigen recognition in MCL and develop targeted therapeutic strategies based on these interactions .
When designing LRPAP1-based B-cell Receptor Antigen Receptor (BAR) bodies for targeted lymphoma therapy, several methodological considerations are essential for optimal efficacy. First, select the appropriate BAR body format based on your therapeutic strategy – options include bispecific constructs (such as anti-CD3/LRPAP1 or anti-CD16/LRPAP1) or antibody-based formats where LRPAP1 replaces variable regions in Fab or IgG1 frameworks . For bispecific constructs, design should include an effector-binding domain (anti-CD3 scFv for T cells or anti-CD16 scFv for NK cells) linked to the precise LRPAP1 epitope region (amino acids 264-318) . When integrating LRPAP1 into antibody formats, test multiple orientations of the epitope region to identify optimal binding configuration – research shows that positioning the MCL-binding epitope at the 5' end of the former variable region (Version A) yields superior binding to LRPAP1-reactive MCL cells . Expression systems significantly impact product quality – prokaryotic systems (E. coli) can be used for initial Fab-format testing, but eukaryotic systems (HEK293 cells) are preferable for full IgG1-format BAR bodies to ensure proper folding and post-translational modifications . Purification protocols should be optimized using appropriate affinity chromatography methods, with careful buffer selection to maintain protein stability. Finally, thorough validation through binding assays (flow cytometry) and functional testing (cytotoxicity assays) using relevant cell lines is critical before advancing to preclinical studies.
Overcoming cross-reactivity issues with LRPAP1 antibodies in complex tissue samples requires a multi-faceted approach. Begin with comprehensive antibody validation using both positive control tissues known to express LRPAP1 and negative control tissues where expression is absent or knocked down. When working with polyclonal LRPAP1 antibodies, which have higher cross-reactivity potential, consider pre-absorption techniques – incubate the antibody with purified LRPAP1 protein or synthetic peptides corresponding to potential cross-reactive epitopes before applying to tissues . Implement stringent blocking procedures using 5-10% normal serum from the same species as the secondary antibody, supplemented with 1-3% BSA to reduce nonspecific binding. Optimize antibody concentration through titration experiments to identify the minimum concentration providing specific signal while minimizing background. For particularly complex samples, consider dual-labeling approaches with antibodies targeting known LRPAP1-associated proteins like LRP1 or LRP2, as co-localization provides stronger evidence of specificity . When analyzing lymphoma samples, include additional markers for B-cell identification to distinguish LRPAP1 expression in malignant versus normal cells. For quantitative applications, establish signal thresholds based on control samples and implement digital image analysis algorithms to distinguish specific from non-specific signals. Finally, validate key findings using complementary approaches such as in situ hybridization for LRPAP1 mRNA or mass spectrometry-based protein identification to provide convergent evidence of specific detection.
Researchers seeking to compare the efficacy of different LRPAP1 BAR body formats in activating immune cells against lymphoma targets should implement a systematic evaluation workflow. Begin with binding assessment through flow cytometry, measuring the affinity of each format (bispecific constructs, Fab-format, and IgG1-format BAR bodies) to both target lymphoma cells (MAVER1, Z138) and effector immune cells (T cells or NK cells) at varying concentrations (1-20 μg/mL) . Quantify binding using mean fluorescence intensity and percentage of positive cells. For functional comparisons, conduct dose-response cytotoxicity assays by co-culturing target lymphoma cells with isolated peripheral blood mononuclear cells (PBMCs) or purified NK cells at various effector-to-target ratios (5:1 to 20:1) in the presence of different BAR body formats at escalating concentrations (1-20 μg/mL) . Measure specific lysis using flow cytometry with viability dyes or chromium release assays. Evaluate immune cell activation by quantifying cytokine production (IFN-γ, TNF-α) via ELISA or cytometric bead arrays and assess activation markers (CD69, CD25) via flow cytometry. Assess off-target effects using control cell lines lacking LRPAP1-reactive BCRs (Granta-519, Mino, U2932) and control BAR bodies with irrelevant targeting domains (e.g., neurabin-I/SAMD14) . Finally, conduct time-course experiments to evaluate the persistence of cytotoxic effects and stability of different formats under physiological conditions. This comprehensive approach enables objective comparison of different BAR body formats to identify optimal candidates for further development.
Producing high-quality recombinant LRPAP1 for antibody validation requires careful consideration of expression systems and purification strategies. For bacterial expression, transform E. coli BL21(DE3) or TG1 strains with expression vectors containing the LRPAP1 coding sequence (full-length or specific epitope regions such as amino acids 264-318) . Induce protein expression with IPTG at concentrations between 0.1-1.0 mM when cultures reach OD600 of 0.6-0.8, and optimize expression by testing different temperatures (16-37°C) and induction times (3-24 hours). For eukaryotic expression, transfect HEK293T cells with mammalian expression vectors using calcium phosphate or lipid-based transfection reagents, and harvest cells after 48-72 hours . For purification of His-tagged LRPAP1, implement immobilized metal affinity chromatography (IMAC) using cobalt-based resins – lyse cells in 10mM TRIS pH 8 buffer for 30 minutes at 4°C, incubate with Talon beads for 30 minutes, wash thoroughly, and elute with 150 mM imidazole before rebuffering in PBS . For FLAG-tagged constructs, use anti-FLAG affinity resins with similar procedures. Further purification can be achieved through size exclusion chromatography to remove aggregates and ensure homogeneity. Validate the purified protein through SDS-PAGE, western blotting, and mass spectrometry to confirm identity and purity. For long-term storage, maintain protein in PBS with 10% glycerol at -80°C, with aliquoting to avoid freeze-thaw cycles that could compromise structural integrity.
When encountering non-specific binding issues with LRPAP1 antibodies in immunohistochemistry, implement a systematic troubleshooting approach. First, optimize tissue fixation and processing – excessive fixation can mask epitopes while insufficient fixation may compromise tissue morphology. For formalin-fixed paraffin-embedded tissues, test multiple antigen retrieval methods including heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) at varying durations (10-30 minutes). Implement a rigorous blocking protocol using 5-10% normal serum from the secondary antibody species, supplemented with 0.1-0.3% Triton X-100 for improved penetration and reduced background . Titrate primary antibody concentrations, testing a range from 1:500 to 1:5000 to identify the optimal signal-to-noise ratio. If high background persists, incorporate additional blocking agents such as avidin/biotin blocking for biotin-based detection systems or 0.1-0.3% hydrogen peroxide to quench endogenous peroxidase activity. Consider overnight incubation at 4°C with primary antibody rather than shorter incubations at room temperature to improve specificity. For difficult tissues, test alternative detection systems such as polymer-based detection instead of avidin-biotin complexes. Incorporate appropriate controls including isotype controls, absorption controls with specific peptides, and tissues known to be negative for LRPAP1 expression. Finally, if problems persist, consider alternative LRPAP1 antibodies targeting different epitopes or switch to fluorescent detection methods which sometimes offer improved signal discrimination.
To rigorously evaluate LRPAP1 antibody specificity across different species, implement a comprehensive experimental design that addresses both sequence homology and functional conservation. Begin with in silico analysis by aligning LRPAP1 protein sequences across target species (human, mouse, rat, and others of interest) to identify conserved regions and potential epitope recognition sites . Select antibodies raised against highly conserved regions if cross-reactivity is desired, or species-specific regions if discrimination is needed. Prepare a panel of positive control samples from each species, including both cell lines and tissue lysates known to express LRPAP1. For western blot validation, run samples from multiple species side-by-side on the same gel to allow direct comparison of band sizes and intensities. Expected molecular weights should be consistent with species variations (approximately 39 kDa for human LRPAP1) . For immunohistochemistry or immunofluorescence validation, process tissues from different species using identical protocols, analyzing both specificity of cellular localization and staining intensity. Include competitive inhibition controls where the antibody is pre-incubated with excess recombinant LRPAP1 protein or immunizing peptide – specific staining should be abolished across all species if the antibody is truly recognizing LRPAP1. For functional validation, test the antibody's ability to immunoprecipitate LRPAP1 from lysates of different species and confirm by mass spectrometry. Finally, validate findings with knockout or knockdown controls in available model systems to confirm absence of signal when LRPAP1 is not expressed.
LRPAP1 antibodies offer significant potential for developing personalized therapies for mantle cell lymphoma (MCL) patients through a multi-phase approach. Initially, implement diagnostic screening using flow cytometry with LRPAP1 antibodies or recombinant LRPAP1 protein to identify patients whose lymphoma B-cell receptors (BCRs) recognize LRPAP1 as an autoantigen . This patient stratification is crucial as only a subset of MCL cases exhibit LRPAP1-reactive BCRs. For patients with LRPAP1-reactive lymphomas, develop personalized BAR body (B-cell Receptor Antigen Receptor) therapies by incorporating patient-specific LRPAP1 epitope regions into therapeutic constructs. The IgG1-format LRPAP1 BAR bodies have demonstrated particular promise, showing dose-dependent cytotoxicity against LRPAP1-reactive MCL cells with lysis rates ranging from 2.5% at 1.0 μg/mL to 43.3% at 20 μg/mL in MAVER1 cells . A comprehensive therapeutic approach would involve combining LRPAP1-targeted constructs with immune effector cells, such as activated NK cells or T cells, in adoptive cell therapy protocols. Monitor treatment response using minimal residual disease assessment with LRPAP1 antibodies to detect remaining lymphoma cells. Furthermore, investigate potential synergies between LRPAP1-targeted therapies and standard treatments (BTK inhibitors, anti-CD20 antibodies) through combination therapy protocols. This personalized medicine approach represents a significant advancement over conventional one-size-fits-all lymphoma treatments by specifically targeting the unique BCR characteristics of individual patients' malignant cells.
LRPAP1 antibodies present valuable tools for investigating neurodegenerative disorders related to lipoprotein receptor dysfunction through multiple research avenues. As LRPAP1 functions as a chaperone for LDL receptor family members including LRP1 and LRP2, which are implicated in Alzheimer's disease and other neurodegenerative conditions, these antibodies can elucidate disease mechanisms . Researchers should implement immunohistochemistry protocols using LRPAP1 antibodies on brain tissue sections from neurodegenerative disease models and patient samples to map expression patterns and potential alterations in disease states. Co-localization studies with antibodies against amyloid-beta, tau, and apolipoprotein E can reveal spatial relationships between LRPAP1 and pathological hallmarks of neurodegeneration. In vitro studies using primary neuronal cultures or brain organoids can employ LRPAP1 antibodies to track receptor trafficking under normal and pathological conditions. For mechanistic investigations, immunoprecipitation with LRPAP1 antibodies followed by mass spectrometry can identify novel interaction partners in the context of neurodegeneration. Functional studies can explore how LRPAP1 modulates lipoprotein receptor-mediated uptake of neurotoxic proteins using antibody-mediated neutralization or detection approaches. Therapeutically, investigate whether modulating LRPAP1-receptor interactions using antibodies or derived fragments could normalize disturbed receptor trafficking in disease models. This research direction could potentially reveal LRPAP1 as a novel target for therapeutic intervention in neurodegenerative conditions characterized by disrupted lipoprotein receptor function.
Resolving contradictory findings between polyclonal and monoclonal antibodies against LRPAP1 requires a systematic analytical approach addressing the fundamental differences between these antibody types. First, perform comprehensive epitope mapping to determine exactly which regions of LRPAP1 are recognized by each antibody – polyclonal antibodies recognize multiple epitopes while monoclonal antibodies target a single epitope, which may explain discrepancies in complex samples . Evaluate antibody specificity through western blotting against recombinant LRPAP1 protein, full cell lysates, and LRPAP1-knockout controls, looking for differences in banding patterns that might indicate off-target binding. Conduct quantitative cross-reactivity testing using peptide arrays or protein microarrays containing LRPAP1 and structurally similar proteins to assess potential off-target interactions. Investigate potential post-translational modifications of LRPAP1 in your experimental system using mass spectrometry, as differently modified forms might be preferentially recognized by different antibodies . Compare antibody performance across multiple detection methods (western blot, immunoprecipitation, immunohistochemistry) to determine if contradictions are technique-specific. If inconsistencies persist, develop validation experiments using orthogonal approaches such as CRISPR/Cas9-mediated LRPAP1 knockout or siRNA-mediated knockdown to establish true expression patterns. Consider additional factors such as lot-to-lot variability in polyclonal antibodies or potential epitope masking in certain conformational states. For critical applications, generate consensus data by using multiple antibodies targeting different LRPAP1 epitopes, focusing on results that are consistent across different antibody types.
The integration of LRPAP1 antibodies into multiplex imaging systems represents a cutting-edge approach for comprehensive cancer research, particularly in studying B-cell malignancies. Researchers should consider multiple advanced platforms, beginning with multiplex immunofluorescence using tyramide signal amplification (TSA), which allows sequential staining with up to 8 antibodies on a single tissue section. In this approach, optimize LRPAP1 antibody dilution and incubation conditions specifically for the TSA system, and pair with antibodies against B-cell markers (CD20, CD79a), lymphoma subtypes (cyclin D1 for MCL), and microenvironment components (T-cells, macrophages) to provide contextual information. For higher dimensionality, implement imaging mass cytometry (IMC) or co-detection by indexing (CODEX) systems, which can simultaneously visualize 40+ markers including LRPAP1 in conjunction with comprehensive tumor and immune panels. These approaches require metal-conjugated LRPAP1 antibodies (typically lanthanide metals for IMC) and careful panel design to avoid signal overlap. For spatial transcriptomics integration, combine LRPAP1 immunodetection with in situ hybridization techniques to correlate protein expression with LRPAP1 mRNA and other relevant transcripts. Develop computational analysis pipelines specifically designed for these multiplex datasets, including cell segmentation algorithms, neighborhood analysis tools, and machine learning approaches to identify spatial patterns and cellular interactions related to LRPAP1 expression. This comprehensive multiplexed visualization enables researchers to understand LRPAP1 biology within the complex tumor microenvironment context, potentially revealing new insights into lymphoma pathogenesis and treatment response mechanisms.
Several emerging technologies stand to significantly enhance the utility of LRPAP1 antibodies in both research and clinical settings. Single-cell proteomics approaches, including cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq), could integrate LRPAP1 antibodies with oligonucleotide tags to simultaneously profile LRPAP1 protein expression and transcriptomes at single-cell resolution in lymphoma samples. Nanobody development targeting LRPAP1 could overcome traditional antibody limitations due to their smaller size, enhanced tissue penetration, and potentially improved specificity for particular epitope regions. For therapeutic applications, the engineering of LRPAP1-directed chimeric antigen receptor (CAR) T-cells represents a logical extension of the BAR body approach, potentially offering more persistent anti-lymphoma activity . Advanced bispecific antibody formats beyond current designs could further enhance the targeting of LRPAP1-reactive malignant B-cells while minimizing off-target effects. High-throughput antibody validation platforms using CRISPR-edited cell lines with modified LRPAP1 expression could standardize antibody characterization across research laboratories. In the diagnostic realm, the development of proximity ligation assays incorporating LRPAP1 antibodies could enable ultrasensitive detection of LRPAP1-BCR interactions in patient samples. Finally, the integration of LRPAP1 antibodies into microfluidic devices or organ-on-chip platforms could facilitate dynamic studies of LRPAP1 function in physiologically relevant systems. These technological advances collectively promise to expand our understanding of LRPAP1 biology and accelerate the translation of LRPAP1-targeted approaches from basic research to clinical applications in lymphoma and potentially other diseases.
The scientific understanding of LRPAP1 is poised for significant evolution based on current research trajectories, with implications spanning basic molecular biology to clinical applications. The discovery of LRPAP1 as a B-cell receptor autoantigen in mantle cell lymphoma has opened entirely new research dimensions beyond its established role as a chaperone for LDL receptor family members . This unexpected immune recognition function suggests LRPAP1 may have broader immunological roles that remain to be elucidated. Research will likely expand to investigate whether LRPAP1 serves as an autoantigen in other B-cell malignancies or autoimmune conditions. The successful development of LRPAP1-based therapeutic constructs for targeting lymphomas may inspire similar approaches for other diseases where LRPAP1 or its receptor partners play critical roles . Structural biology studies employing cryo-electron microscopy and X-ray crystallography will likely provide detailed insights into how LRPAP1 interacts with LDL receptor family members and potentially with B-cell receptors, enabling structure-based drug design. Systems biology approaches integrating genomics, proteomics, and metabolomics data may reveal previously unrecognized LRPAP1 functions in cellular signaling networks. The role of LRPAP1 in neurodegenerative disorders will likely receive increased attention given the involvement of its partner receptors in conditions like Alzheimer's disease . Investigation of LRPAP1 polymorphisms and expression variations across populations may uncover connections to disease susceptibility or progression. Finally, the therapeutic applications of LRPAP1-targeting approaches will likely expand beyond current concepts, potentially including combination therapies with established agents and extension to other malignancies where LRPAP1 biology intersects with disease mechanisms.
Optimizing LRPAP1 antibody performance across various experimental applications requires careful attention to multiple parameters. The following table provides detailed optimization guidelines for achieving reliable results:
| Application | Sample Preparation | Antibody Dilution Range | Incubation Conditions | Detection System | Critical Controls | Troubleshooting Tips |
|---|---|---|---|---|---|---|
| Western Blot | 10% SDS-PAGE, PVDF membrane | 1:1000-1:5000 | 1 hour at RT or overnight at 4°C | HRP-conjugated secondary antibody, chemiluminescence | Recombinant LRPAP1, LRPAP1-knockout cells | Use 5% milk or BSA in TBST for blocking; avoid detergents that may disrupt epitopes |
| Immunohistochemistry | FFPE tissues, 5μm sections, citrate buffer (pH 6.0) antigen retrieval | 1:100-1:500 | 1-2 hours at RT or overnight at 4°C | DAB detection system | Known positive tissues, peptide competition | Optimize antigen retrieval time; use avidin-biotin blocking for high background |
| Immunofluorescence | 4% PFA fixation, 0.2% Triton X-100 permeabilization | 1:200-1:1000 | Overnight at 4°C | Fluorophore-conjugated secondary antibodies | Peptide competition, LRPAP1-knockout cells | Increase blocking time for high background; use mounting media with anti-fade properties |
| Flow Cytometry | 5 × 10^6 cells, gentle fixation | 5-10 μg/mL | 30 minutes at 4°C | APC or PE-conjugated secondary antibody | LRPAP1+ and LRPAP1- cell lines | Avoid enzymatic dissociation methods that might cleave surface epitopes |
| ELISA | Coat plates with 1-5 μg/mL recombinant LRPAP1 | 1:500-1:2000 | 1-2 hours at RT | HRP substrate with colorimetric detection | Standard curve, blocking peptides | Optimize coating buffer pH; use carrier proteins to minimize non-specific binding |
| Immunoprecipitation | Lysis in RIPA or gentler NP-40 buffer | 2-5 μg per 500 μg protein | Overnight at 4°C | Protein A/G beads | Pre-immune serum, IgG control | Add protease inhibitors freshly; avoid harsh detergents that disrupt protein-protein interactions |