The antibody is validated for:
Western blot: Detects LNPEP in HeLa lysates (1 µg/mL) and human brain/kidney tissues .
Immunohistochemistry: Stains human heart/kidney tissues (antigen retrieval with TE buffer) .
LNPEP (Q9UIQ6) is a 1025-amino-acid type II transmembrane glycoprotein:
Biological roles:
Recent studies (2023) using bioinformatics tools (TCGA, TIMER2.0) revealed:
Ovarian cancer (OV): High LNPEP expression correlates with poor prognosis (Cox regression, P=0.009) .
Immune infiltration: Positively associated with Th1/Th2 cells, macrophages, and immunomodulators (LinkedOmic) .
From the Human Protein Atlas :
LNPEP is highly expressed in:
Neurological tissues: Hippocampus, amygdala, basal ganglia.
Endocrine organs: Adrenal glands, pituitary gland.
Gastrointestinal tract: Stomach, small intestine.
LNPEP’s dual role in RAS and immune modulation positions it as a therapeutic target for:
R&D Systems (LNPEP Antibody AF6386)
Frontiers in Immunology (Multifaceted Role of LNPEP)
PMC/NCBI (LNPEP in Ovarian Cancer)
Proteintech (LNPEP Antibody 55154-1-AP)
Human Protein Atlas (LNPEP Expression)
LNPEP (Leucyl/cystinyl Aminopeptidase) is a zinc-dependent aminopeptidase involved in peptide hormone regulation, antigen presentation, and cell surface processing. Commercial antibodies target multiple epitope regions across the protein, enabling researchers to select antibodies appropriate for specific experimental needs. The most common commercial antibodies target regions including the N-terminus (AA 1-110, AA 23-52, AA 16-46), middle sections, and C-terminus (AA 900-950) . One well-characterized antibody (ABIN6143284) targets amino acids 1-110 with the sequence "MEPFTNDRLQ LPRNMIENSM FEEEPDVVDL AKEPCLHPLE PDEVEYEPRG SRLLVRGLGE HEMEEDEEDY ESSAKLLGMS FMNRSSGLRN SATGYRQSPD GACSVPSART" . These diverse targeting options allow researchers to select antibodies specific to domains of interest or regions without post-translational modifications.
| Antibody Target Region | Host | Type | Common Applications |
|---|---|---|---|
| AA 1-110 (N-Term) | Rabbit | Polyclonal | WB, IHC |
| AA 23-52 (N-Term) | Rabbit | Polyclonal | WB, FACS, IHC(p) |
| AA 16-46 (N-Term) | Rabbit | Polyclonal | WB, FACS, IHC(p), EIA |
| AA 926-1025 | Mouse | Polyclonal | WB, ELISA |
| C-Term (AA 900-950) | Rabbit | Polyclonal | WB, ELISA, IF |
| Full length (AA 1-1025) | Mouse | Polyclonal | WB |
LNPEP antibodies are validated for multiple experimental applications, with each antibody optimized for specific techniques. Western Blotting (WB) remains the most common application, with most commercial antibodies showing reliable detection of LNPEP at its expected molecular weight (~160-170 kDa) . Immunohistochemistry (IHC) applications include both paraffin-embedded (IHC-P) and frozen sections, with antibodies like ABIN6143284 specifically validated for these techniques . Flow cytometry (FACS) is supported by several antibodies targeting N-terminal regions (AA 23-52, AA 16-46), which can detect cell surface-expressed LNPEP . Additional validated applications include ELISA (particularly for antibodies targeting AA 926-1025 and AA 900-950), immunoprecipitation (IP), and immunofluorescence (IF) . When selecting a LNPEP antibody, researchers should verify validation data for their specific application of interest.
Species cross-reactivity is a critical consideration when selecting LNPEP antibodies, especially for comparative studies or research using animal models. Most commercially available LNPEP antibodies demonstrate reactivity against human LNPEP, while a subset also cross-react with mouse and rat orthologs . For example, the LNPEP antibody targeting amino acids 1-110 (ABIN6143284) shows verified cross-reactivity with human, mouse, and rat samples . When working with less common model organisms, cross-reactivity must be empirically determined as it often cannot be reliably predicted from sequence homology alone. Researchers should carefully review cross-reactivity data and, when possible, validate with positive control samples from their species of interest. For evolutionarily distant species or when cross-reactivity data is unavailable, preliminary validation experiments should be conducted before proceeding with full-scale studies.
Proper storage and handling of LNPEP antibodies are essential for maintaining their functionality and extending their usable lifespan. Most LNPEP antibodies should be stored at -20°C for long-term preservation, with working aliquots kept at 4°C for up to one month to minimize freeze-thaw cycles . Repeated freeze-thaw cycles dramatically reduce antibody activity and should be avoided by preparing appropriate single-use aliquots before freezing. Working dilutions should be prepared fresh on the day of use in appropriate buffers (typically PBS or TBS with 0.1% BSA); these dilutions should not be stored for future use. Many antibody preparations contain preservatives like sodium azide that may interfere with certain applications, particularly those involving HRP-conjugated reagents. When shipping or receiving antibodies, cold chain maintenance is critical—upon receipt, antibodies should be immediately stored at recommended temperatures and centrifuged briefly before opening to collect contents at the bottom of the vial.
Epitope targeting significantly impacts antibody utility for specific research questions and should guide selection based on experimental goals. For studying full-length LNPEP, antibodies targeting conserved N-terminal regions (AA 1-110) provide reliable detection across multiple applications . When investigating LNPEP processing or specific isoforms, C-terminal antibodies (e.g., those targeting AA 900-950) may better distinguish between processed forms . Researchers studying protein-protein interactions should select antibodies targeting regions distinct from known interaction domains to avoid competitive binding that could yield false negative results. For detection of active LNPEP, antibodies targeting regions outside the catalytic domain are preferable to avoid interference with enzymatic activity. When conducting inhibition studies, antibodies recognizing epitopes within functional domains may serve as blocking reagents. Additionally, researchers should consider whether their application requires detection of native (conformational) or denatured (linear) epitopes, as antibodies often show preference for one state over the other .
High-resolution epitope characterization enhances experimental design and interpretation when working with LNPEP antibodies. High-density peptide microarrays represent a cutting-edge approach for comprehensive epitope mapping . This technology allows synthesis of all possible overlapping peptides (typically 15-mers) spanning the entire LNPEP sequence on a single microscope slide containing more than 200,000 peptide fields . Single-residue substitution analysis, where each position in potential epitopes is systematically replaced with all 20 natural amino acids, reveals critical binding residues at single-amino acid resolution . Statistical approaches including ANOVA followed by post-hoc tests like Tukey's Honest Significance Difference (HSD) identify positions statistically significant for antibody binding . Position-specific scoring matrices (PSSMs) quantify each residue's contribution to binding affinity, with Rq values (ratio of obtained to critical studentized range values) providing statistical measures of significance . Cross-reactivity potential can be assessed by including peptides from homologous proteins (e.g., BSA, RSA) in the array .
Rigorous controls are essential for generating reliable and interpretable data with LNPEP antibodies across all applications. For Western blotting, positive controls should include recombinant LNPEP or lysates from tissues known to express high levels of LNPEP (e.g., placenta for human studies) . Negative controls should include LNPEP-knockout or knockdown samples when available; alternatively, tissues known not to express LNPEP can serve this purpose. For immunostaining techniques (IHC/IF), primary antibody omission controls assess non-specific binding of secondary reagents, while isotype controls (using non-targeting antibodies of the same isotype, e.g., rabbit IgG for rabbit polyclonal LNPEP antibodies) evaluate non-specific binding due to antibody class . Peptide competition controls, where the antibody is pre-incubated with excess immunizing peptide before application, confirm epitope specificity. For quantitative applications like ELISA or flow cytometry, standard curves with recombinant protein determine the linear dynamic range. Finally, biological replicates and technical replicates help distinguish experimental variation from true biological effects.
Cross-reactivity presents significant challenges for LNPEP antibody applications but can be systematically addressed through multiple approaches. Sequence analysis using bioinformatics tools can identify proteins with sequence similarity to LNPEP epitopes, predicting potential cross-reactants. Genetic validation using LNPEP knockout or knockdown samples provides the gold standard for specificity verification—if signals persist in these samples, cross-reactivity is likely present. Peptide competition assays, where antibodies are pre-incubated with excess immunizing peptide, can distinguish specific from non-specific binding; signals that remain after peptide blocking likely represent cross-reactivity . The multi-antibody approach, using several antibodies targeting different LNPEP epitopes, can corroborate findings and identify discrepancies suggesting cross-reactivity. High-density peptide arrays provide comprehensive cross-reactivity assessment by including peptides from potential cross-reactive proteins . For example, including bovine serum albumin (BSA) peptides in arrays revealed moderate cross-reaction with some LNPEP antibodies, while no cross-reactivity was observed with rabbit serum albumin peptides .
Multiple factors contribute to variability in LNPEP detection, requiring systematic mitigation strategies. Antibody characteristics significantly impact consistency—polyclonal antibodies like those commonly used for LNPEP detection show batch-to-batch variation that can be addressed by purchasing larger lots for long-term studies or by validating new lots against reference standards . Sample preparation variability can be reduced through standardized protocols with precisely timed fixation (for IHC/IF) or lysis conditions (for WB/IP) that don't compromise epitope integrity. LNPEP's variable expression levels across tissues and cell types necessitate optimized protein loading and detection methods; for low-expression samples, more sensitive detection systems (e.g., amplification systems for IHC or enhanced chemiluminescence for WB) may be required. Post-translational modifications of LNPEP can mask epitopes or alter antibody affinity, necessitating careful selection of antibodies targeting regions without known modifications. Protocol consistency is paramount—standardized incubation times, temperatures, buffer compositions, and washing steps minimize technical variability. Finally, quantitative analysis should include appropriate normalization to loading controls or reference genes.
An optimized Western blotting protocol for LNPEP detection requires careful attention to each step. For sample preparation, cells or tissues should be lysed in RIPA buffer supplemented with protease inhibitors, with 20-50 μg total protein loaded per lane . LNPEP's large molecular weight (~160-170 kDa) necessitates low percentage (7-8%) SDS-PAGE gels and extended running times to achieve optimal separation. Transfer to PVDF membranes (preferred over nitrocellulose for high molecular weight proteins) should use wet transfer systems at 30V overnight at 4°C to ensure complete transfer of the large protein. Blocking should occur in 5% non-fat dry milk in TBST for 1 hour at room temperature, though BSA may be substituted when detecting phosphorylated forms. Primary antibody incubation should follow manufacturer's recommended dilutions (typically 1:500 to 1:2000 for LNPEP antibodies) in blocking buffer overnight at 4°C . After washing 4-5 times with TBST (5 minutes each), membranes should be incubated with appropriate HRP-conjugated secondary antibody (typically 1:5000) for 1 hour at room temperature. Following additional washes, enhanced chemiluminescence detection with extended exposure times (1-5 minutes) often yields optimal results for LNPEP visualization.
Optimizing immunohistochemistry for LNPEP detection requires careful attention to fixation, antigen retrieval, and detection methods. Tissue fixation should use 10% neutral buffered formalin for 24-48 hours, as overfixation can mask LNPEP epitopes while underfixation compromises tissue morphology . Paraffin-embedded tissues should be sectioned at 4-6 μm thickness for optimal antibody penetration. Antigen retrieval is critical—heat-induced epitope retrieval using citrate buffer (pH 6.0) in a pressure cooker (20 minutes) typically works well for LNPEP antibodies targeting N-terminal regions, while EDTA buffer (pH 9.0) may better expose C-terminal epitopes . Endogenous peroxidase blocking with 3% H₂O₂ (10 minutes) and protein blocking with serum-free protein block (20 minutes) reduce background. Primary antibody incubation should use optimized dilutions (typically 1:100 to 1:500 for LNPEP antibodies) overnight at 4°C in a humidity chamber . Polymer-based detection systems generally provide better sensitivity for polyclonal LNPEP antibodies than biotin-based methods. DAB development time should be standardized (typically 3-5 minutes) across experiments, and hematoxylin counterstaining should be light to avoid obscuring specific LNPEP staining.
Comprehensive validation of new LNPEP antibodies should follow a multi-tiered approach to ensure reliability across intended applications. Western blot validation should confirm detection of a single band at the expected molecular weight (~160-170 kDa) in positive control samples (placenta, kidney, or LNPEP-transfected cells) . Specificity validation should include genetic approaches (testing on LNPEP knockout/knockdown samples) and peptide competition assays where pre-incubation with immunizing peptide abolishes specific signal . Application-specific validation is essential—antibodies performing well in Western blots may not function for immunoprecipitation or immunostaining. For IHC/IF applications, staining patterns should be compared with known LNPEP subcellular localization patterns in the literature. Cross-species validation requires testing with samples from each species of interest rather than assuming cross-reactivity based on sequence homology alone . Quantitative applications require establishing a linear dynamic range using dilution series of recombinant protein or lysates. Finally, validation data should be thoroughly documented according to guidelines from the International Working Group for Antibody Validation, including images of full blots, positive and negative controls, and detailed methodological information.
When encountering weak or inconsistent LNPEP antibody signals, a systematic troubleshooting approach should address multiple potential factors. For Western blotting, increasing protein loading (50-100 μg), extending primary antibody incubation (overnight at 4°C), and using more sensitive detection reagents (e.g., enhanced chemiluminescence plus systems) can improve signal strength . For immunostaining applications, optimizing antigen retrieval is often critical—testing multiple methods (heat-induced versus enzymatic) and buffer systems (citrate pH 6.0 versus EDTA pH 9.0) can dramatically improve epitope accessibility . Signal amplification systems (tyramide signal amplification or polymer-based detection) can enhance sensitivity for low-abundance LNPEP detection. For all applications, antibody concentration should be systematically titrated, as both too-dilute and too-concentrated antibodies can yield suboptimal results. Fresh antibody aliquots should be tested if degradation is suspected. Sample-related issues can be addressed by verifying LNPEP expression using RT-PCR, ensuring appropriate sample handling (fresh preparation, adequate protease inhibitors), and using positive control samples known to express LNPEP. Finally, switching to alternative antibodies targeting different LNPEP epitopes may resolve persistent detection problems, as some epitopes may be masked by post-translational modifications or protein interactions in specific experimental contexts .
Selecting and optimizing LNPEP antibodies for co-immunoprecipitation (co-IP) requires specific considerations distinct from other applications. Antibody selection should prioritize those that recognize native LNPEP—this information is sometimes provided by manufacturers but often requires empirical testing . Epitope accessibility in the native state is critical; antibodies targeting regions that remain exposed in the folded protein (often N-terminal or C-terminal domains) typically perform better than those targeting potentially buried regions . Pre-clearing lysates with protein A/G beads before adding antibodies reduces non-specific binding. Antibody amounts require optimization—typically 2-5 μg per 500 μg of total protein, with too much antibody potentially causing high background and too little resulting in inefficient precipitation . Lysis conditions are crucial for co-IP success; gentler non-ionic detergents (NP-40, Triton X-100 at 0.5-1%) better preserve protein-protein interactions than stronger ionic detergents (SDS, deoxycholate). Buffer salt concentration affects interaction stability—physiological concentrations (150 mM NaCl) generally work well, but some interactions may require lower salt conditions. Validation should include immunoprecipitation with non-specific IgG controls and reciprocal co-IP (immunoprecipitating the interaction partner and blotting for LNPEP) to confirm specificity of interactions.
Emerging antibody technologies offer promising avenues to advance LNPEP research beyond current limitations. Single-domain antibodies (nanobodies) derived from camelid heavy-chain antibodies provide superior access to sterically hindered epitopes due to their smaller size, potentially revealing currently inaccessible functional domains of LNPEP . Recombinant antibody engineering allows creation of bispecific antibodies simultaneously targeting LNPEP and interaction partners, enabling novel approaches to study protein complexes. Site-specific conjugation technologies permit precise attachment of fluorophores or other labels at defined positions, minimizing interference with antigen recognition. Antibody fragments like Fab and scFv offer improved tissue penetration for imaging applications and reduced non-specific binding through Fc regions. Intrabodies—antibodies designed for intracellular expression—could enable visualization or manipulation of LNPEP in living cells. High-throughput epitope binning using technologies like array-based surface plasmon resonance can rapidly characterize large panels of LNPEP antibodies, identifying those with non-overlapping epitopes for sandwich assays. These emerging technologies will likely facilitate more precise interrogation of LNPEP biology, particularly for studying its diverse cellular functions and protein-protein interactions in native contexts.
Multi-color imaging with LNPEP antibodies requires careful planning to achieve reliable co-localization data. Primary antibody selection should prioritize different host species for LNPEP and other target proteins (e.g., rabbit anti-LNPEP with mouse anti-protein X) to avoid cross-reactivity between secondary antibodies . When this is not possible, directly conjugated primary antibodies or sequential staining protocols can circumvent cross-reactivity issues. Spectral considerations are critical—fluorophore selection should minimize spectral overlap, and when overlap is unavoidable, appropriate compensation controls should be included. Sequential scanning rather than simultaneous acquisition can further reduce bleed-through artifacts. Controls should include single-color samples for compensation settings and fluorescence-minus-one (FMO) controls to establish gating boundaries. For super-resolution microscopy applications, antibody-fluorophore combinations should be optimized for photostability and brightness, with direct conjugation often preferred to minimize the distance between epitope and fluorophore. For live-cell applications, non-perturbing binding is essential—antibody fragments (Fab, scFv) typically cause less clustering or internalization than full IgG. Finally, quantitative co-localization analysis should employ appropriate statistical methods (Pearson's correlation, Manders' coefficients) rather than relying solely on visual assessment of overlap.