NXPH2 Antibody is a primary antibody designed to detect and study the Neurexophilin-2 (NXPH2) protein, a member of the Neurexophilin family. These proteins are secreted signaling molecules that bind to α-neurexins and other receptors, influencing neurotransmitter release and cellular adhesion . The antibody is primarily used in immunohistochemistry (IHC), Western blot (WB), and ELISA to analyze NXPH2 expression in tissues, cells, and biological fluids.
NXPH2 Antibodies are typically rabbit polyclonal, unconjugated, and affinity-purified for high specificity. Key features include:
NXPH2 Antibodies are critical for studying:
Neurological and Kidney Functions: NXPH2 is highly expressed in brain and kidney .
Cancer Biology: IHC studies show varied expression in cancers (e.g., colorectal, breast, prostate, lung) .
Immune Responses: GWAS linked NXPH2 SNPs (e.g., rs62163199) to HLA class I alloantibody formation in women, suggesting a role in immune regulation .
NXPH2 Antibodies undergo rigorous validation:
Tissue Arrays: Tested on 44 normal and 20 cancer tissues to confirm specificity .
Protein Arrays: Cross-reactivity evaluated against 364 human recombinant proteins .
Predictive Models: Sequence alignment predicts reactivity in non-human species (e.g., Pig, Horse) .
Immune Regulation: GWAS implicated NXPH2 in HLA alloimmunization, with SNPs reducing alloantibody risk (OR=0.51) .
Protein Expression: Secreted NXPH2 localizes to extracellular regions, consistent with its role as a signaling molecule .
Cancer Prognosis: High NXPH2 expression in certain cancers (e.g., breast, lung) may correlate with survival outcomes (Kaplan-Meier plots available via Human Protein Atlas) .
NXPH2 (Neurexophilin-2) is a secreted protein belonging to the neurexophilin family. It functions as a signaling molecule that resembles neuropeptides and acts by binding to alpha-neurexins and possibly other receptors . NXPH2 is primarily expressed in brain and kidney tissues , making it relevant for neuroscience research investigating synaptic function and neuronal signaling pathways.
The importance of NXPH2 in neuroscience stems from its potential role in modulating synaptic transmission through its interaction with neurexins, which are key synaptic adhesion molecules. Researching NXPH2 can provide insights into neural circuit formation, synaptic plasticity, and potentially neurological disorders associated with synaptic dysfunction.
NXPH2 is a relatively small protein with a calculated molecular weight of approximately 30 kDa . The protein contains several key structural features:
A signal peptide for secretion
An N-terminal non-conserved domain
A central conserved domain characteristic of the neurexophilin family
Notable post-translational modifications of NXPH2 include:
| Site | PTM Type | Source |
|---|---|---|
| T131 | Phosphorylation | Uniprot |
| Y252 | Phosphorylation | Uniprot |
| Y254 | Phosphorylation | Uniprot |
| Y260 | Phosphorylation | Uniprot |
NXPH2 undergoes proteolytic processing at the boundary between the N-terminal non-conserved domain and the central conserved domain, particularly in neuron-like cells . This processing is likely important for its biological function.
Distinguishing between NXPH2 (Neurexophilin-2) and NXPE2 (Neurexophilin and PC-esterase domain family member 2) requires attention to several key differences:
Molecular Weight: NXPH2 has a molecular weight of approximately 30 kDa , while NXPE2 is significantly larger at 64.9 kDa with 559 amino acid residues .
Subcellular Localization: NXPH2 is primarily a secreted protein , whereas NXPE2 is localized to the membrane .
Tissue Expression Pattern: While NXPH2 is predominantly expressed in brain and kidney , NXPE2 shows notable expression in salivary gland, epididymis, colon, and appendix .
Antibody Validation: When using antibodies, researchers should verify target specificity through techniques such as Western blotting at the expected molecular weight, using appropriate positive and negative controls, and confirming the expression pattern matches known tissue distribution.
Domain Structure: The proteins belong to different families despite similarity in naming, with NXPE2 containing a PC-esterase domain not present in NXPH2.
Optimizing Western blot conditions for NXPH2 detection requires careful consideration of several parameters:
Sample Preparation:
Antibody Selection and Dilution:
Electrophoresis and Transfer Conditions:
Blocking and Detection:
5% non-fat milk or BSA in TBST is typically sufficient for blocking
Incubate primary antibody at 4°C overnight for optimal binding
HRP-conjugated secondary antibodies with compatible chemiluminescent detection systems provide good sensitivity
Controls and Validation:
Include both positive (brain/kidney tissue lysates) and negative controls
Consider using recombinant NXPH2 as a standard
Expected results should show a distinct band at approximately 30 kDa, though additional bands may appear if detecting proteolytically processed forms of the protein.
Optimizing immunohistochemistry (IHC) for NXPH2 detection in brain tissues requires attention to several critical parameters:
Tissue Preparation:
For IHC-P: Use 4% paraformaldehyde fixation (4-24 hours depending on tissue size)
For IHC-F: Snap-freeze tissue in OCT compound and prepare 10-20 μm sections
Consider antigen retrieval methods (citrate buffer pH 6.0 or EDTA buffer pH 9.0) for formalin-fixed tissues
Antibody Selection and Dilution:
Protocol Optimization:
Extended blocking (1-2 hours) with 5-10% normal serum from the same species as the secondary antibody
Optimize primary antibody incubation time (overnight at 4°C often yields best results)
Include 0.1-0.3% Triton X-100 in buffers to enhance antibody penetration for neuron-specific staining
Controls and Validation:
Positive controls: Human or mouse brain sections (regions with known NXPH2 expression)
Negative controls: Omission of primary antibody and tissues with minimal NXPH2 expression
Consider co-staining with neuronal markers to confirm cell-type specificity
Signal Enhancement and Background Reduction:
Tyramide signal amplification for low-abundance targets
Autofluorescence quenching for brain tissues (Sudan Black B treatment)
Careful washing steps (at least 3×10 minutes) after antibody incubations
Expected results should show predominantly neuronal staining patterns in brain tissues with potential secretory patterns consistent with NXPH2's role as a secreted protein.
The selection between polyclonal and monoclonal NXPH2 antibodies should be guided by experimental requirements and research objectives:
Polyclonal NXPH2 Antibodies:
Advantages:
Recognize multiple epitopes, potentially increasing detection sensitivity
Better tolerance for protein denaturation or conformational changes
Generally less expensive and easier to produce
May detect various isoforms or post-translationally modified variants
Best Applications:
Limitations:
Batch-to-batch variability requiring validation across lots
Potential for cross-reactivity with related proteins
Less specificity for distinguishing highly similar proteins
Monoclonal NXPH2 Antibodies:
Advantages:
Consistent specificity across experiments and batches
Reduced background and non-specific binding
Superior for distinguishing between closely related proteins
Better for quantitative applications
Best Applications:
Experiments requiring high reproducibility
Flow cytometry and immunoprecipitation
Therapeutic and diagnostic applications
Long-term studies requiring consistent reagents
Limitations:
May recognize only a single epitope, reducing detection if that epitope is masked
Sometimes less sensitive than polyclonal antibodies
More susceptible to epitope loss through fixation or denaturation
Generally more expensive
For discovery-phase research, polyclonal antibodies may provide better detection
For specific quantitative assays or advanced applications, monoclonal antibodies offer greater consistency
Consider using both types for validation and confirmation of findings
Non-specific binding and high background are common challenges when working with NXPH2 antibodies. These methodological approaches can help address these issues:
Antibody Dilution Optimization:
Blocking Optimization:
Test different blocking reagents (BSA, normal serum, commercial blocking buffers)
Extend blocking time to 1-2 hours at room temperature
Consider adding 0.1-0.5% Tween-20 to blocking buffer to reduce hydrophobic interactions
For tissue sections, include avidin/biotin blocking steps if using biotinylated detection systems
Washing Procedures:
Increase washing duration (3-5 washes of 5-10 minutes each)
Use gentle agitation during washing steps
Consider adding higher salt concentration (up to 500 mM NaCl) to wash buffer for reducing ionic interactions
Secondary Antibody Considerations:
Pre-adsorb secondary antibodies against tissue powder
Use secondary antibodies specifically cross-adsorbed against other species
Reduce secondary antibody concentration
Sample-Specific Approaches:
For Western blot: Use freshly prepared samples and PVDF membranes (better for protein binding)
For IHC: Optimize fixation time and antigen retrieval methods
For ICC: Test different fixation protocols (4% PFA vs. methanol)
Controls to Include:
No primary antibody control to assess secondary antibody background
Isotype control to evaluate non-specific binding
Pre-absorption with immunizing peptide to confirm specificity
If high background persists, consider switching to a different NXPH2 antibody targeting an alternative epitope or from a different manufacturer.
When NXPH2 antibodies reveal unexpected molecular weight bands in Western blots, a systematic investigation approach is necessary:
Verification of Expected NXPH2 Molecular Weight:
Analysis of Higher Molecular Weight Bands:
Potential protein aggregation: Include reducing agents (DTT or β-mercaptoethanol) in sample buffer
Possible dimers or multimers: Try more stringent denaturation conditions
Glycosylation or other post-translational modifications: Consider enzymatic treatment (e.g., PNGase F for N-linked glycans)
Evaluate cross-reactivity with related neurexophilin family members
Investigation of Lower Molecular Weight Bands:
Potential proteolytic processing: NXPH2 may be processed at the boundary between domains
Degradation products: Add additional protease inhibitors during sample preparation
Alternative splice variants: Verify against known transcript variants
C-terminal or N-terminal fragments: Use antibodies targeting different regions of NXPH2
Experimental Validation Approaches:
Peptide competition assay: Pre-incubate antibody with immunizing peptide to identify specific bands
Molecular weight calibration: Use precise molecular weight standards
Knockdown/knockout validation: Compare samples with reduced or eliminated NXPH2 expression
Immunoprecipitation followed by mass spectrometry to identify protein components
Technical Optimization:
Adjust gel percentage to better resolve proteins in the range of interest
Optimize transfer conditions for efficient protein transfer
Consider gradient gels for better separation across a wide molecular weight range
Documentation and Reporting:
Clearly document all bands observed and their reproducibility
Compare results with published literature on NXPH2
Contact antibody manufacturers for technical support and additional validation data
Batch-to-batch variability is a significant challenge, particularly with polyclonal NXPH2 antibodies. These strategies can help address inconsistencies:
Comprehensive Validation of New Antibody Batches:
Perform side-by-side comparison with previous batches
Test multiple applications (WB, IHC, IF) to assess performance across platforms
Document dilution optimization for each new batch
Create a standardized validation protocol specific to your experimental needs
Reference Sample Archive:
Maintain frozen aliquots of standard samples (brain/kidney lysates) for batch testing
Use recombinant NXPH2 protein as a consistent positive control
Create a reference image library of expected results for comparison
Quantitative Assessment:
Measure signal-to-noise ratios across batches
Quantify detection sensitivity using dilution series of positive controls
Document epitope-specific reactivity using peptide arrays if available
Antibody Storage and Handling Optimization:
Alternative Strategies:
Purchase larger antibody lots when possible to reduce batch changes
Consider switching to monoclonal antibodies for greater consistency
Validate multiple antibodies targeting different epitopes of NXPH2
Implement multiplexed detection approaches combining different NXPH2 antibodies
Documentation and Quality Control:
Maintain detailed records of antibody lot numbers and performance
Develop standard operating procedures (SOPs) for antibody validation
Consider implementing a rating system for antibody performance across applications
By implementing these practices, researchers can significantly reduce the impact of batch-to-batch variability on experimental outcomes and maintain consistency in NXPH2 detection.
Computational approaches are increasingly valuable for designing and optimizing NXPH2-specific antibodies, enabling researchers to move beyond traditional selection methods:
Epitope Prediction and Selection:
Analyze NXPH2 sequence for regions of high antigenicity and surface accessibility
Identify conserved regions across species for cross-reactive antibodies
Select epitopes distinct from other neurexophilin family members to ensure specificity
Target regions less likely to undergo post-translational modifications
Structure-Based Antibody Design:
Utilize protein structure prediction tools to model NXPH2 3D conformation
Apply molecular docking simulations to optimize antibody-antigen interactions
Identify key binding residues for rational mutation to enhance affinity
Implement advanced systems like JAM for de novo antibody design with precise epitope targeting
Machine Learning Applications:
Train models on existing antibody-antigen datasets to predict binding properties
Apply deep learning approaches to optimize complementarity-determining regions (CDRs)
Use sequence-based predictions to identify frameworks with optimal expression characteristics
Implement models that disentangle different binding modes for improved specificity
Library Design and Screening Optimization:
Design natural diversity libraries based on somatic hypermutation patterns
Use computational approaches to create focused libraries with higher hit rates
Implement in silico screening before experimental validation
Apply bacterial display systems to evaluate expression and stability in parallel with binding properties
Specificity Engineering:
Identify key residues that distinguish NXPH2 from other neurexophilin family members
Model cross-reactivity potential against similar proteins
Design antibodies with customized specificity profiles for either high specificity or controlled cross-reactivity
Apply computational tools for negative design against unwanted interactions
Experimental Validation of Computational Predictions:
Implement phage display with high-throughput sequencing for experimental validation
Test computationally designed variants with customized specificity profiles
Validate predictions through in vitro binding assays and structural studies
Iterate between computational predictions and experimental results
These computational approaches not only improve antibody specificity and affinity but can also enhance expression yields and stability, making them valuable tools for developing improved NXPH2 antibodies for research and potential therapeutic applications.
Studying the interactions between NXPH2 and alpha-neurexins requires sophisticated methodological approaches spanning from biochemical to cellular analyses:
In Vitro Binding Assays:
Surface Plasmon Resonance (SPR): Measure real-time binding kinetics (Kon, Koff) and affinity (KD) between purified NXPH2 and alpha-neurexins
Microscale Thermophoresis (MST): Assess binding in solution with minimal protein consumption
Bio-Layer Interferometry (BLI): Analyze binding without microfluidics requirements
ELISA-Based Interaction Assays: Develop quantitative sandwich assays using NXPH2 antibodies
Co-Immunoprecipitation Approaches:
Use NXPH2 antibodies to pull down protein complexes from neural tissues or cell cultures
Perform reciprocal co-immunoprecipitation with alpha-neurexin antibodies
Couple with mass spectrometry for unbiased identification of the complete interactome
Apply chemical crosslinking to stabilize transient interactions
Structural Biology Methodologies:
X-ray Crystallography: Determine atomic-resolution structures of NXPH2-neurexin complexes
Cryo-Electron Microscopy: Visualize larger complexes without crystallization requirements
NMR Spectroscopy: Map binding interfaces through chemical shift perturbations
Hydrogen-Deuterium Exchange Mass Spectrometry: Identify regions involved in binding
Cell-Based Interaction Systems:
Proximity Ligation Assay (PLA): Visualize interactions in situ with subcellular resolution
FRET/BRET Approaches: Measure protein-protein interactions in living cells
Split Reporter Systems: GFP complementation or luciferase-based approaches
Cell Surface Binding Assays: Using recombinant proteins to identify binding to membrane-expressed partners
Functional Modulation Experiments:
Use NXPH2 antibodies to block interactions with alpha-neurexins
Apply gene knockout/knockdown approaches to assess functional consequences
Develop peptide mimetics of binding interfaces for competitive inhibition
Perform domain swapping or mutagenesis to identify critical binding determinants
Advanced Imaging Methodologies:
Super-Resolution Microscopy: Visualize co-localization at synaptic structures
Single-Molecule Tracking: Follow dynamics of NXPH2-neurexin interactions
Expansion Microscopy: Achieve enhanced spatial resolution in tissue preparations
Correlative Light and Electron Microscopy: Combine functional and ultrastructural data
These complementary approaches provide a comprehensive understanding of NXPH2-alpha-neurexin interactions, from molecular binding mechanisms to functional consequences at synapses.
Comprehensive validation of NXPH2 antibody specificity across platforms requires a multi-dimensional approach:
Orthogonal Validation Strategies:
Genetic Validation: Test antibodies in tissues/cells with NXPH2 gene knockout or knockdown
Peptide Competition: Pre-absorption with immunizing peptide should abolish specific signal
Multiple Antibody Concordance: Compare results from antibodies targeting different NXPH2 epitopes
Recombinant Protein Controls: Use purified NXPH2 as positive control in multiple assays
Cross-Platform Technical Validation:
Validate in Western blot to confirm molecular weight specificity (expected ~30 kDa)
Verify tissue expression pattern in IHC matches known distribution (brain, kidney)
Confirm subcellular localization in IF/ICC consistent with secreted protein characteristics
Assess specificity in immunoprecipitation followed by mass spectrometry
Cross-Species Reactivity Assessment:
Specificity Against Related Proteins:
Test for cross-reactivity with other neurexophilin family members (NXPH1, NXPH3, NXPH4)
Assess potential cross-reactivity with NXPE family proteins
Perform immunoblotting against recombinant related proteins
Consider heterologous expression systems overexpressing target or related proteins
Advanced Validation Technologies:
Epitope Mapping: Identify precise binding sites using peptide arrays or hydrogen-deuterium exchange
Immunodepletion Studies: Sequential immunoprecipitation to demonstrate antibody specificity
Multiple Detection Methods: Validate using different secondary antibody systems
Super-Resolution Microscopy: Confirm expected localization patterns with nanoscale precision
Documentation and Reporting Standards:
This comprehensive validation strategy ensures that experimental results with NXPH2 antibodies are robust, reproducible, and truly reflect the biological properties of NXPH2 rather than technical artifacts.
Optimizing NXPH2 antibody stability and expression requires consideration of multiple parameters throughout the antibody production and handling processes:
Framework Optimization for Enhanced Expression:
Leverage naturally occurring somatic hypermutation diversity to identify stabilizing frameworks
Use bacterial antibody display (BAD) systems to screen variants for improved expression
Identify framework residues that influence expression levels through systematic mutation
Apply computational approaches to predict stability-enhancing modifications
Production System Selection and Optimization:
Compare expression levels between prokaryotic (E. coli) and eukaryotic systems (mammalian cells, insect cells)
Optimize codon usage for the selected expression host
Evaluate different signal peptides for improved secretion
Test various induction conditions and expression temperatures
Purification Strategy Optimization:
Implement affinity chromatography with protein A/G for IgG formats
Consider ion exchange chromatography as a polishing step
Use size exclusion chromatography to remove aggregates
Optimize buffer conditions during purification to maintain stability
Formulation for Enhanced Stability:
Storage and Handling Optimization:
Stability Enhancement Through Protein Engineering:
Identify and remove hydrophobic patches prone to aggregation
Engineer disulfide bonds for increased thermostability
Remove deamidation-prone asparagine residues
Consider humanization approaches for improved stability
Validation of Optimization Success:
Assess thermostability through differential scanning fluorimetry
Monitor accelerated stability studies under stress conditions
Evaluate long-term activity retention through functional assays
Test freeze-thaw stability over multiple cycles
Designing experiments to investigate NXPH2's role in neurological function using antibody-based approaches requires integrating multiple methodologies:
Expression Mapping in Neural Systems:
Comprehensive Brain Region Analysis: Map NXPH2 expression across brain regions using IHC with validated antibodies
Developmental Time Course: Track expression changes during neural development
Cell-Type Specificity: Combine with neuronal, glial, and synaptic markers for co-localization studies
Subcellular Localization: Use immunogold electron microscopy for precise synaptic localization
Functional Perturbation Studies:
Acute Neutralization: Apply function-blocking NXPH2 antibodies in electrophysiological recordings
Chronic Interference: Introduce antibodies to neuronal cultures for long-term functional assessment
In Vivo Applications: Use intracerebroventricular antibody delivery for behavioral studies
Ex Vivo Preparations: Apply antibodies to brain slices for circuit-level analysis
Protein-Protein Interaction Analysis:
Synaptic Interactome Mapping: Use NXPH2 antibodies for immunoprecipitation coupled with mass spectrometry
In Situ Interaction Visualization: Apply proximity ligation assays to visualize NXPH2-neurexin interactions
Competition Studies: Use soluble NXPH2 and anti-NXPH2 antibodies to disrupt native interactions
Activity-Dependent Dynamics: Analyze how neural activity modulates NXPH2 interactions
Pathophysiological Relevance Assessment:
Disease Model Evaluation: Compare NXPH2 expression and localization in neurological disorder models
Human Tissue Studies: Examine NXPH2 patterns in post-mortem samples from patients with neurological conditions
Biomarker Potential: Develop sensitive ELISA using NXPH2 antibodies for CSF or serum detection
Therapeutic Exploration: Test antibody-based modulation of NXPH2 function in disease models
Advanced Imaging Approaches:
Live Imaging: Use fluorescently-labeled antibody fragments to track NXPH2 dynamics
Super-Resolution Microscopy: Resolve NXPH2 distribution at synapses with nanoscale precision
Array Tomography: Combine ultrathin sectioning with immunofluorescence for 3D reconstruction
Expansion Microscopy: Achieve enhanced spatial resolution in complex neural tissues
Molecular Mechanism Dissection:
Post-Translational Modification Analysis: Use modification-specific antibodies to detect NXPH2 phosphorylation
Proteolytic Processing Studies: Develop antibodies specific to different NXPH2 domains to track processing
Secretion Pathway Analysis: Track NXPH2 through secretory compartments using compartment markers
Receptor Binding and Signaling: Monitor downstream signaling events following NXPH2-neurexin interaction
These experimental approaches provide a comprehensive framework for understanding NXPH2's neurological functions, from molecular mechanisms to circuit-level roles, potentially revealing new insights into synaptic function and neurological disorders.
The following table summarizes key characteristics of commercially available NXPH2 antibodies based on the provided search results:
| Characteristic | Boster Bio (A16657) | Uping Bio (YP-mAb-05826) | Affinity Biosciences (DF9683) | Bioss (bs-11171r-bf555) |
|---|---|---|---|---|
| Antibody Type | Polyclonal | Monoclonal | Polyclonal | Polyclonal |
| Host Species | Rabbit | Mouse | Rabbit | Rabbit |
| Reactivity | Human, Mouse, Rat | Human, Mouse | Human, Mouse | Human, Mouse, Rat, others |
| Applications | WB | WB, ELISA | WB, IHC | WB, IF(IHC-P), IF(IHC-F), IF(ICC) |
| Dilution Range (WB) | 1:500-1:1000 | 1:500-2000 | 1:500-2000 | 1:300-5000 |
| Immunogen | Synthetic peptide from Human NXPH2 | Peptide (AA range: 140-220) | Not specified | KLH-conjugated synthetic peptide (range: 151-250/264) |
| Form | Liquid | Liquid | Liquid | Liquid |
| Storage | -20°C (1 year), 4°C (1 month) | -20°C (1 year) | -20°C | -20°C |
| Special Features | Validated in WB | IgG isotype | Application in IHC | Conjugated to AbBy Fluor® 555 |
| Expected MW | ~30 kDa | 29 kDa | 30 kDa | Not specified |
This comparison allows researchers to select the most appropriate NXPH2 antibody based on their specific experimental requirements, considering factors such as application needs, species reactivity, and available detection systems.
The following table outlines standardized protocol parameters for common NXPH2 antibody applications: