LRRFIP2 (Leucine-Rich Repeat FLI-I-Interacting Protein 2) antibodies are specialized immunological tools designed to detect and study the LRRFIP2 protein, a key regulator of inflammatory and signaling pathways. These antibodies are primarily used in research to analyze protein expression, localization, and interactions in diverse biological contexts, including inflammation, Wnt signaling, and cancer progression. Below is a structured overview of their applications, specifications, and research implications.
LRRFIP2 antibodies enable precise analysis of the protein’s role in cellular processes:
Purpose: Quantify LRRFIP2 expression levels in lysates.
Key Findings:
Purpose: Localize LRRFIP2 within cells.
Key Findings:
Purpose: Study protein-protein interactions.
Key Findings:
LRRFIP2 antibodies have elucidated critical roles of the protein in inflammation and signaling:
Mechanism: LRRFIP2 inhibits NLRP3 inflammasome activation by recruiting Flightless-I to caspase-1, suppressing IL-1β production .
Evidence:
Role: LRRFIP2 activates Wnt signaling by interacting with Dishevelled (DVL3), enhancing β-catenin stability .
Evidence:
Role: LRRFIP2 positively regulates TLR4-NF-κB signaling by competing with Flightless-I for MYD88 binding .
Evidence:
Role: LRRFIP2 isoforms (e.g., variant 2 vs. 3) differentially regulate gastric cancer metastasis. Antibodies detect isoform-specific expression .
Evidence:
LRRFIP2 (Leucine-rich repeat flightless-interacting protein 2) is a protein with a canonical length of 721 amino acid residues and a mass of 82.2 kDa in humans. It functions as an activator of the canonical Wnt signaling pathway, acting in association with DVL3 upstream of CTNNB1/beta-catenin. LRRFIP2 is widely expressed throughout the body, with highest expression levels observed in heart and skeletal muscle tissues . Recent research has also identified LRRFIP2 as a negative regulator of the NLRP3 inflammasome, indicating its importance in regulating inflammatory responses . This dual functionality makes LRRFIP2 a significant target for researchers studying both developmental biology through Wnt signaling and inflammatory disease mechanisms through inflammasome regulation.
Commercial LRRFIP2 antibodies demonstrate variable species reactivity profiles depending on the specific product. Based on available data, many LRRFIP2 antibodies show reactivity toward human (Homo sapiens) samples, with some cross-reacting with mouse (Ms) and rat (Rt) LRRFIP2 . This cross-reactivity is supported by the evolutionary conservation of LRRFIP2, with orthologs identified in multiple species including mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken . When selecting an antibody for experiments involving non-human species, researchers should carefully verify the documented species reactivity and consider performing validation experiments to confirm cross-reactivity in their specific experimental system.
LRRFIP2 antibodies are utilized across several experimental techniques, with Western Blot being the most common application. Additional validated applications include:
Western Blotting (WB): For detecting LRRFIP2 protein expression levels and molecular weight
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative measurement of LRRFIP2
Immunohistochemistry (IHC): For visualization of LRRFIP2 in tissue sections
Immunocytochemistry (ICC): For cellular localization studies
Immunofluorescence (IF): For subcellular localization and co-localization studies
Working dilutions vary by application and specific antibody product, typically ranging from 1:100 for immunocytochemistry to 1:1,000 for Western blotting applications . Researchers should optimize dilutions for their specific experimental conditions.
LRRFIP2 has been identified as a negative regulator of NLRP3 inflammasome activation through co-immunoprecipitation and mass spectrometry studies. To investigate this regulatory mechanism, researchers can employ the following approach:
Co-immunoprecipitation protocol: Use anti-NLRP3 antibody with total cell lysates of macrophages primed with lipopolysaccharide (LPS) and then stimulated with ATP to identify LRRFIP2 as an interacting partner .
Temporal interaction analysis: LRRFIP2 co-precipitates with NLRP3 approximately 10 minutes after ATP stimulation or 30 minutes after Nigericin stimulation, suggesting stimulus-specific timing for this interaction .
Domain mapping: N-terminal and coil motifs of LRRFIP2 are required for its inhibitory effect on NLRP3 inflammasome activation. Researchers can use truncated mutants of LRRFIP2 to identify which domains are necessary for its regulatory function .
Functional validation: Silencing LRRFIP2 in macrophages enhances NLRP3 inflammasome-mediated caspase-1 cleavage and IL-1β production, while overexpression of LRRFIP2 reduces these inflammatory outputs .
This methodological approach allows researchers to thoroughly investigate LRRFIP2's role in regulating inflammatory responses mediated by the NLRP3 inflammasome.
To investigate LRRFIP2's role as an activator of the canonical Wnt signaling pathway, researchers can implement the following experimental design:
Expression analysis: Use LRRFIP2 antibodies for Western blotting to determine basal expression levels across various cell types with known Wnt pathway activity.
Gain/loss-of-function experiments:
Overexpress LRRFIP2 using expression vectors and confirm using LRRFIP2 antibodies via Western blot
Silence LRRFIP2 using siRNA technology and verify knockdown efficiency
Pathway activation assessment:
Quantify β-catenin nuclear translocation using nuclear/cytoplasmic fractionation followed by Western blotting
Measure Wnt target gene expression by qRT-PCR
Employ TOP/FOP flash reporter assays to measure TCF/LEF-dependent transcription
Protein interaction studies:
Perform co-immunoprecipitation with LRRFIP2 antibodies to detect interaction with DVL3
Use proximity ligation assays to visualize LRRFIP2-DVL3 interactions in situ
Domain mapping: Create truncated constructs of LRRFIP2 to identify which domains are essential for Wnt pathway activation, similar to approaches used in inflammasome studies .
This comprehensive experimental approach enables researchers to establish both the necessity and sufficiency of LRRFIP2 in Wnt signaling activation and define its precise mechanistic role in the pathway.
For investigating LRRFIP2's role in inflammatory disease models, researchers can employ the following methodological approach:
In vivo silencing: Utilize siRNA technology to silence LRRFIP2 expression in mouse models. This has been demonstrated to enhance NLRP3 inflammasome activation and increase inflammatory cell recruitment in peritonitis models .
Tissue analysis protocol:
Collect peritoneal exudate cells (PECs) after inflammatory challenge
Perform flow cytometry to quantify total inflammatory cell recruitment
Specifically analyze neutrophil and Ly6C+ monocyte populations
Use LRRFIP2 antibodies for immunohistochemistry to assess expression patterns in inflamed tissues
Cytokine profiling:
Measure IL-1β production using ELISA in tissue samples and culture supernatants
Assess other inflammatory cytokines to understand broader effects
Cellular mechanism studies:
This methodological framework allows researchers to establish the functional significance of LRRFIP2 in inflammatory disease processes while providing mechanistic insights into its regulatory role.
For optimal Western blot detection of LRRFIP2, researchers should consider the following protocol guidelines:
Sample preparation:
Cell lysis buffer: RIPA buffer supplemented with protease inhibitors
Protein concentration: 20-40 μg total protein per lane
Denaturation: 95°C for 5 minutes in Laemmli buffer with β-mercaptoethanol
Gel electrophoresis parameters:
Gel percentage: 8-10% SDS-PAGE (due to LRRFIP2's 82.2 kDa size)
Running conditions: 100V through stacking gel, 120V through resolving gel
Transfer conditions:
Wet transfer recommended: 100V for 90 minutes or 30V overnight at 4°C
PVDF membrane preferred over nitrocellulose for higher sensitivity
Antibody incubation:
Detection system:
Validation controls:
This optimized protocol significantly enhances detection specificity and sensitivity for LRRFIP2 in Western blotting applications.
When performing immunohistochemistry (IHC) or immunofluorescence (IF) with LRRFIP2 antibodies, researchers should consider these methodological aspects:
Fixation method comparison:
| Fixation Method | Advantages | Disadvantages |
|---|---|---|
| 4% Paraformaldehyde | Preserves morphology, compatible with most antibodies | May mask some epitopes |
| Methanol | Better for nuclear antigens, enhances some epitope accessibility | Can distort membrane proteins |
| Acetone | Good for cytoskeletal proteins | Poor morphological preservation |
Antigen retrieval considerations:
Heat-induced epitope retrieval (HIER): Citrate buffer (pH 6.0) for 20 minutes
Enzymatic retrieval: Consider proteinase K treatment if HIER is ineffective
Antibody optimization:
Signal detection systems:
IHC: DAB (3,3'-diaminobenzidine) substrate for permanent staining
IF: Alexa Fluor conjugates (488, 555, 647) for fluorescence detection
Counterstaining options:
IHC: Hematoxylin for nuclear visualization
IF: DAPI for nuclear counterstaining, phalloidin for F-actin visualization
Controls:
These methodological considerations ensure optimal detection of LRRFIP2 in tissue and cellular samples while minimizing background and non-specific staining.
For successful co-immunoprecipitation (co-IP) experiments involving LRRFIP2, researchers should consider these technical aspects:
Lysis buffer optimization:
For protein-protein interactions: Non-denaturing buffers (e.g., 1% NP-40, 150 mM NaCl, 50 mM Tris-HCl pH 8.0)
Protease inhibitor cocktail: Essential to prevent degradation
Phosphatase inhibitors: Important when studying signaling pathways
Pre-clearing protocol:
Incubate lysates with Protein A/G beads for 1 hour at 4°C
Remove beads by centrifugation before adding the specific antibody
Antibody amounts and ratios:
LRRFIP2 immunoprecipitation: 2-5 μg antibody per 500 μg total protein
For co-IP with NLRP3: Similar amounts to maintain stoichiometric balance
Incubation conditions:
Antibody binding: 2-4 hours or overnight at 4°C with gentle rotation
Protein A/G bead binding: 1-2 hours at 4°C with gentle rotation
Washing stringency optimization:
Less stringent: PBS with 0.1% Tween-20
More stringent: Increasing salt concentration (150-500 mM NaCl)
Number of washes: 3-5 times, 5 minutes each
Elution methods:
Denaturing: SDS sample buffer at 95°C for 5 minutes
Non-denaturing: Glycine buffer (pH 2.5) followed by neutralization
Detection strategy:
Timing considerations:
This detailed co-IP protocol enhances the detection of physiologically relevant LRRFIP2 protein interactions while minimizing non-specific binding.
Researchers often encounter these challenges when detecting LRRFIP2 by Western blot:
When facing persistent issues, researchers may consider:
Using recombinant LRRFIP2 protein as a positive control
Comparing results with different LRRFIP2 antibody clones
Validating with knockdown/knockout samples if available
Optimizing transfer conditions for proteins >80 kDa
These troubleshooting approaches address the most common technical challenges in LRRFIP2 Western blot detection.
To address specificity concerns with LRRFIP2 antibodies, researchers should implement the following validation strategies:
Genetic validation approaches:
siRNA/shRNA knockdown: Compare antibody signal in control vs. LRRFIP2-depleted samples
CRISPR/Cas9 knockout: Generate complete LRRFIP2 knockout for definitive negative control
Overexpression: Transfect cells with LRRFIP2 expression constructs to confirm signal increase
Epitope competition assays:
Pre-incubate antibody with immunizing peptide (if available)
Compare signal with and without peptide competition
A specific antibody should show significant signal reduction after peptide competition
Multiple antibody validation:
Use antibodies targeting different LRRFIP2 epitopes
Compare staining/binding patterns across antibodies
Consistent patterns across antibodies suggest specificity
Cross-reactivity testing:
Test antibody in species with known LRRFIP2 homology
Perform Western blot on lysates from various species
Results should align with expected cross-reactivity based on epitope conservation
Biophysical validation:
Application-specific controls:
For ICC/IF: Include secondary-only controls
For IHC: Use isotype controls at matching concentrations
For IP: Include IgG control immunoprecipitations
These comprehensive validation approaches ensure that experimental observations genuinely reflect LRRFIP2 biology rather than antibody artifacts.
When researchers encounter conflicting results between different LRRFIP2 antibodies, these systematic approaches can help reconcile discrepancies:
Epitope mapping analysis:
Determine the exact epitopes recognized by each antibody
Epitopes in highly conserved regions may yield more consistent results
Epitopes in regions subject to alternative splicing may detect different isoforms
Isoform-specific detection:
Post-translational modification interference:
Some epitopes may be masked by phosphorylation, glycosylation, or other modifications
Treatment with phosphatases or glycosidases can reveal if modifications affect antibody binding
Consider antibodies specifically targeting modified forms if relevant
Context-dependent expression:
Methodological validation:
Perform side-by-side comparisons using identical protocols
Systematically vary antibody concentrations to determine optimal working ranges
Consider cell/tissue-specific optimization
Reconciliation strategy:
For multiple positive signals: Accept overlapping patterns as likely true positives
For contradictory signals: Prioritize antibodies with genetic validation
Consider implementing orthogonal detection methods (e.g., mass spectrometry)
This systematic approach helps researchers determine which antibody is most appropriate for their specific experimental context and biological question.
Several emerging applications of LRRFIP2 antibodies show promise for advancing inflammation research:
In vivo imaging applications:
Development of near-infrared fluorophore-conjugated LRRFIP2 antibodies for non-invasive imaging
Tracking LRRFIP2 expression dynamics during inflammatory disease progression
Potential for monitoring therapeutic efficacy in real-time
Single-cell analysis techniques:
Adapting LRRFIP2 antibodies for mass cytometry (CyTOF) applications
Single-cell Western blotting to detect LRRFIP2 in rare cell populations
Incorporating LRRFIP2 into multiparameter flow cytometry panels for immune cell profiling
Spatial transcriptomics integration:
Combining LRRFIP2 antibody staining with spatial transcriptomics
Correlating protein expression with localized gene expression patterns
Creating tissue atlases of LRRFIP2 expression in inflammatory diseases
Therapeutic target validation:
Using LRRFIP2 antibodies to validate small molecule modulators of LRRFIP2 function
Developing blocking antibodies to manipulate LRRFIP2-mediated inflammasome regulation
Screening for compounds that enhance LRRFIP2's anti-inflammatory properties
Biomarker development:
Investigating LRRFIP2 expression patterns in inflammatory diseases
Correlating LRRFIP2 levels with disease severity and therapeutic responses
Developing standardized assays for clinical research applications
These emerging applications leverage LRRFIP2's role as a negative regulator of NLRP3 inflammasome activation , potentially opening new avenues for therapeutic intervention in inflammatory diseases.
LRRFIP2 antibodies can provide unique insights into the cross-talk between Wnt signaling and inflammation pathways through these methodological approaches:
Dual pathway visualization:
Multiplex immunofluorescence combining LRRFIP2 with both Wnt pathway markers (β-catenin, TCF/LEF) and inflammasome components (NLRP3, ASC)
Super-resolution microscopy to visualize nanoscale protein interactions
Live-cell imaging to track dynamic interactions during pathway activation
Protein complex analysis:
Sequential co-immunoprecipitation to identify multi-protein complexes
Blue native PAGE to preserve native protein complexes for Western blot analysis
Proximity-dependent labeling approaches (BioID, APEX) with LRRFIP2 as bait
Cellular response mapping:
Stimulate cells with both Wnt ligands and inflammasome activators
Use LRRFIP2 antibodies to track redistribution and modification states
Perform time-course analyses to determine sequence of events in dual pathway activation
Context-dependent interactions:
Compare LRRFIP2 interaction partners in different cell types (epithelial vs. immune cells)
Analyze interaction networks under homeostatic vs. inflammatory conditions
Investigate tissue-specific LRRFIP2 functions in models of inflammatory disease
Mechanistic dissection:
Use domain-specific LRRFIP2 antibodies to determine which regions mediate Wnt vs. inflammasome interactions
Employ conformation-specific antibodies to detect activation states
Develop phospho-specific antibodies if regulatory phosphorylation sites are identified
These approaches can help elucidate how LRRFIP2 might integrate or segregate signals between developmental Wnt pathways and inflammatory response pathways, potentially revealing new therapeutic targets at this intersection.
Emerging methodologies for studying LRRFIP2 in macrophage polarization and function include:
Single-cell analysis protocols:
Single-cell RNA-seq combined with LRRFIP2 protein detection (CITE-seq)
Mass cytometry (CyTOF) panels incorporating LRRFIP2 antibodies
Correlation of LRRFIP2 levels with macrophage polarization markers (M1 vs. M2)
Advanced imaging techniques:
Lattice light-sheet microscopy for long-term tracking of LRRFIP2 dynamics during polarization
Multi-color STORM or PALM super-resolution imaging to visualize nanoscale protein complexes
Correlative light and electron microscopy (CLEM) to connect LRRFIP2 localization with ultrastructural features
Functional genomics approaches:
CRISPR-Cas9 screening to identify genes that modify LRRFIP2 function in macrophages
CRISPRi/CRISPRa for tunable modulation of LRRFIP2 expression
CRISPR base editing to introduce specific mutations for structure-function studies
Macrophage-specific in vivo models:
Development of macrophage-specific LRRFIP2 conditional knockout mice
Adoptive transfer studies with LRRFIP2-modified macrophages
Tissue-specific deletion to examine organ-specific functions
Secretome analysis:
Quantitative proteomics of macrophage secretomes after LRRFIP2 manipulation
Cytokine profiling comparing wild-type and LRRFIP2-deficient macrophages
Extracellular vesicle characterization to identify LRRFIP2-dependent secretory pathways
These methodologies will provide deeper insights into how LRRFIP2 influences macrophage polarization states and effector functions, particularly in the context of its role as a negative regulator of NLRP3 inflammasome activation .