The HRP-conjugated RHBDF2 antibody (e.g., ABIN1930520) is a polyclonal rabbit antibody raised against a synthetic peptide spanning amino acids 80–109 in the N-terminal region of human RHBDF2 . Key features include:
Parameter | Details |
---|---|
Epitope | N-terminal region (AA 80–109) |
Host | Rabbit |
Conjugate | Horseradish Peroxidase (HRP) |
Reactivity | Human |
Applications | ELISA, Western Blotting (WB), Immunohistochemistry (IHC) |
Purification | Affinity-purified via peptide chromatography |
Molecular Weight Target | ~90–98 kDa (varies by isoform and post-translational modifications) |
The HRP-conjugated antibody enables direct detection of RHBDF2 in lysates without requiring secondary antibodies. For example:
Detection in MCF-7 breast cancer cells: A 1:500 dilution of ABIN1930520 identified RHBDF2 at ~98 kDa in MCF-7 lysates, confirmed by chemiluminescence .
Validation in knockout models: In mouse bone marrow macrophage fractions, the antibody distinguished wild-type RHBDF2 from knockout (KO) samples, confirming specificity .
HRP-conjugated antibodies are suitable for IHC due to their compatibility with chromogenic substrates (e.g., DAB). ABIN1930520 has been used to localize RHBDF2 in human tissues, though specific IHC protocols (e.g., antigen retrieval methods) may require optimization .
HRP-conjugated antibodies simplify ELISA workflows. ABIN1930520 detects RHBDF2 in human serum or cell culture supernatants, enabling quantification of protein levels in inflammatory or pathological contexts .
NAFLD/NASH: RHBDF2 regulates MAP3K7 phosphorylation and inflammatory signaling, contributing to steatohepatitis. Its degradation via the ubiquitin-proteasome system is modulated by E3 ligases like Trim31 .
Familial Esophageal Cancer: Gain-of-function mutations in RHBDF2 (e.g., c.557T>C) cause tylosis with esophageal cancer syndrome by hyperactivating EGFR signaling .
Protein Stability: HRP-conjugated antibodies have been used to track RHBDF2 degradation in cycloheximide-treated hepatocytes, confirming its proteasomal turnover .
ADAM17 Regulation: RHBDF2 antibodies (e.g., MAB10048) help study the pseudoprotease’s role in shedding soluble ligands like TNF-α and CD40, critical in inflammation .
Dilution Optimization: Working concentrations vary by application (e.g., 1:500 for WB, 1:50–1:500 for IHC) .
Cross-Reactivity: Polyclonal antibodies (e.g., ABIN1930520) may require blocking with non-specific IgG to reduce background noise in IHC or ELISA .
Storage: HRP-conjugated antibodies are typically stored at 4°C (short-term) or -20°C (long-term) to preserve enzyme activity .
RHBDF2 encodes the protein 'rhomboid 5 homolog 2' in humans, also known by alternative names including RHBDL6, TOC, RHBDL5, TEC, inactive rhomboid protein 2, and rhomboid family member 2. This approximately 96.7 kilodalton protein has gained significant research interest due to its identification as a late-onset Alzheimer's disease risk factor . More specifically, RHBDF2 (also called iRhom2) functions as a modifier of microglial TREM2 proteolysis, establishing its importance in neurological disease pathways . Research utilizing RHBDF2 antibodies is essential for understanding its role in both physiological processes and pathological conditions, particularly in neurodegenerative disorders.
RHBDF2 antibodies vary in their cross-reactivity profiles, with many commercial options showing reactivity to human RHBDF2, while a subset also recognizes mouse, rat, canine, porcine, or monkey orthologs . When planning experiments, researchers must carefully evaluate whether the antibody has been validated for their specific model organism. Cross-reactivity validation should involve Western blot analysis with positive and negative controls for each species. While sequence homology can suggest potential cross-reactivity, empirical validation is essential as small differences in epitope regions can significantly impact antibody performance across species, potentially leading to false negative results or misleading data interpretation.
Optimization of HRP-conjugated RHBDF2 antibody dilutions for Western blotting requires systematic titration across multiple cell types. Begin with the manufacturer's recommended dilution range (typically 1:1,000 for many RHBDF2 antibodies) and perform a dilution series . For cells with known high RHBDF2 expression (e.g., HaCaT and A549 cell lines), testing dilutions from 1:500 to 1:2,000 may be appropriate . For other cell types, particularly primary cells or those with lower expression levels, more concentrated antibody solutions (1:250 to 1:500) may be necessary. Critical optimization parameters include:
Cell Type | Starting Dilution | Optimization Range | Protein Loading | Incubation Time |
---|---|---|---|---|
HaCaT | 1:1,000 | 1:500 - 1:2,000 | 20-30 μg | 1-2 hours or overnight at 4°C |
A549 | 1:1,000 | 1:500 - 1:2,000 | 20-30 μg | 1-2 hours or overnight at 4°C |
BV2 | 1:500 | 1:250 - 1:1,000 | 30-50 μg | 2 hours or overnight at 4°C |
Primary microglia | 1:250 | 1:100 - 1:500 | 40-60 μg | Overnight at 4°C |
Validation should include positive and negative controls, and optimization may require adjustment of blocking conditions and washing stringency to minimize background while maintaining specific signal detection .
Investigation of RHBDF2-ADAM17 interactions requires multiple complementary approaches. Co-immunoprecipitation using HRP-conjugated RHBDF2 antibodies can identify protein-protein interactions, while knockout validation through CRISPR-Cas9 provides essential controls . For CRISPR-based approaches targeting RHBDF2, researchers can use validated primer sequences such as forward guide #1: 5′-GCCCACACCGTATCTGTTCT-3′, reverse guide #1: 5′-CTGCAAGAGATGTGGGTGAA-3′; forward guide #2: 5′-CAGGAACCCAGGGCTTTAGG-3′, reverse guide #2: 5′-TTCTGGCCTTCCACATCCAC-3′ . For secretome analysis of RHBDF2-dependent ADAM17 substrates, the high-performance secretome protein enrichment with click sugars (hiSPECS) method can be employed to identify membrane proteins undergoing proteolysis in wild-type versus RHBDF2 knockout conditions . This approach allows detection of low-abundance secreted proteins and shed membrane protein ectodomains while removing abundant serum proteins like albumin from the analysis .
Flow cytometry provides a powerful tool for quantifying cell surface protein levels affected by RHBDF2 activity. For example, to quantify TREM2 surface levels in relation to RHBDF2/iRhom2 function, cells should be seeded at appropriate density (e.g., 5 × 10^5 cells in 12-well plates) and harvested by gentle detachment to preserve surface epitopes . Critical methodology includes:
Pre-incubation with blocking buffer (PBS, 1% BSA, 0.25% NaN₃) containing 1% mouse SeroBlock FcR to prevent non-specific binding
Staining with fluorophore-conjugated antibodies against the target protein (e.g., APC-conjugated anti-TREM2) with appropriate isotype controls
Multiple washing steps (minimum three washes) to remove unbound antibody
Exclusion of doublets and dead cells through proper gating and viability dyes (e.g., propidium iodide)
Collection of sufficient events (minimum 10,000 per sample) for statistical analysis
This approach allows quantitative assessment of how RHBDF2 modulates surface expression of target proteins, providing insights into its regulatory functions in protein trafficking and shedding.
High background is a common challenge when using HRP-conjugated antibodies in immunohistochemistry. For RHBDF2 detection in tissues like human kidney and small intestine, where this antibody has been validated, several optimization strategies can minimize background while preserving specific signal :
Optimize antigen retrieval conditions by testing multiple buffers (citrate pH 6.0 vs. EDTA pH 9.0) and retrieval durations
Implement dual blocking with both protein blocking (3-5% BSA or normal serum) and peroxidase blocking (3% hydrogen peroxide for 10-15 minutes)
Increase washing stringency with PBS-T (0.1-0.3% Tween-20) and extend washing times
Optimize antibody dilution (starting at 1:100 as recommended but testing 1:50-1:200 range)
Reduce substrate development time and monitor microscopically to prevent overdevelopment
Include absorption controls by pre-incubating the antibody with recombinant RHBDF2 protein
For multi-labeling experiments, tyramide signal amplification can enhance sensitivity while maintaining specificity, allowing detection of low-abundance epitopes without increasing background.
Rigorous validation of RHBDF2 antibody specificity is essential for generating reliable research data. A comprehensive validation approach should include:
CRISPR/Cas9 knockout validation: Generate RHBDF2 knockout cell lines using validated guide RNAs and confirm editing efficiency using Synthego's Inference of CRISPR Edits tool (https://ice.synthego.com/)[2]
siRNA/shRNA knockdown: Perform transient and stable knockdown experiments with at least two different siRNA/shRNA constructs targeting different regions of RHBDF2
Western blot analysis: Compare protein expression between wild-type and knockout/knockdown samples across multiple cell types, looking for absence or significant reduction of the expected ~96.7 kDa band
Immunocytochemistry comparison: Perform parallel staining of wild-type and knockout cells to confirm loss of specific signal
Recombinant protein controls: Use purified recombinant RHBDF2 protein as a positive control in Western blot applications
Cross-reactivity assessment: Test for potential cross-reactivity with related rhomboid family proteins, particularly RHBDF1, which shares structural similarity with RHBDF2
This multi-faceted validation approach ensures that experimental findings can be confidently attributed to specific RHBDF2 detection rather than antibody cross-reactivity or non-specific binding.
ELISA protocols for quantifying RHBDF2-regulated proteins, such as shed TREM2, require careful optimization. Based on established methodologies:
Sample preparation: Culture supernatants should be diluted appropriately (e.g., 1:40 in PBS with 0.05% Tween, 1% BSA, and protease inhibitor cocktail) and 50 μl added to each well
Standard curve generation: Create a standard curve using recombinant protein (e.g., for TREM2, use a twofold serial dilution of recombinant mTREM2-FC from 400 pg/ml to 12.5 pg/ml)
Denaturation considerations: For certain proteins with Fc tags, denaturation may be necessary to prevent dimerization (use denaturing buffer containing 200 mM Tris-HCL, pH 6.8, 4% SDS, 40% glycerol, 2% ß-ME, 50 mM EDTA followed by heating at 95°C for 5 min)
Antibody incubation: Incubate samples and standards for 2 hours at room temperature, followed by detection antibody incubation (e.g., rat anti-TREM2 at 1:1,000 dilution) for 1 hour
Signal detection: For HRP-conjugated detection, use appropriate substrate and measure using colorimetric or chemiluminescent detection systems
Data analysis: Apply a four-parameter logistic fit curve regression model for accurate concentration determination
This approach enables precise quantification of soluble factors regulated by RHBDF2, facilitating investigation of its role in protein shedding and secretion pathways.
Discrepancies between protein-level detection using RHBDF2 antibodies and mRNA expression data from transcriptomic studies require careful analysis. Several factors can contribute to such discrepancies:
Post-transcriptional regulation: RHBDF2 may undergo extensive post-transcriptional regulation, including miRNA targeting, resulting in poor correlation between mRNA and protein levels
Protein stability differences: Variations in protein half-life across different tissues or conditions can lead to accumulation of protein despite low mRNA expression
Antibody specificity issues: The antibody may detect cross-reactive epitopes or post-translationally modified forms of RHBDF2 not reflected in transcriptomic data
Splice variant detection: Different antibodies may preferentially detect specific splice variants that are not distinguished in bulk RNA sequencing
To resolve such conflicts, researchers should:
Employ multiple antibodies targeting different epitopes
Validate with orthogonal methods like mass spectrometry
Use reporter systems with epitope tags to track expression
Perform single-cell analyses to identify cell-type-specific expression patterns that may be masked in bulk analyses
Given RHBDF2/iRhom2's identification as a late-onset Alzheimer's disease risk factor and modifier of microglial TREM2 proteolysis , effective experimental designs should incorporate:
Cell models: Compare RHBDF2 function in:
Microglial cell lines (BV2, HMC3)
Primary microglia from wild-type and RHBDF2 knockout mice
iPSC-derived microglia from individuals with and without Alzheimer's risk alleles
Animal models: Utilize:
RHBDF2 knockout mice crossed with Alzheimer's disease models (APP/PS1, 5xFAD)
Conditional/inducible RHBDF2 knockout to distinguish developmental from acute effects
Age-dependent analyses to capture late-onset disease relevance
Human samples:
Post-mortem brain tissue from Alzheimer's patients and controls
Analysis of RHBDF2 expression in relation to disease stage and TREM2 processing
Mechanistic assays:
This multi-level approach addresses both mechanistic questions about RHBDF2 function and translational relevance to Alzheimer's disease pathology.
Integration of RHBDF2 antibody-generated data into multi-omics frameworks requires strategic experimental design and analytical approaches:
Parallel omics sampling: Design experiments to collect samples for antibody-based detection alongside RNA-seq, proteomics, and other omics measurements from the same biological specimens
Normalization strategies: Develop appropriate normalization methods to compare antibody-based quantification with proteomics and transcriptomics data
Pathway enrichment: Conduct pathway analyses incorporating RHBDF2 interactors and substrates identified by antibody-based methods
Temporal profiling: Perform time-course experiments to capture dynamic relationships between RHBDF2-mediated protein shedding and downstream transcriptional responses
An effective integration framework might include:
Antibody-based spatial proteomics to localize RHBDF2
Cell surface proteomics to identify RHBDF2-dependent membrane protein changes
Phosphoproteomics to capture signaling events downstream of RHBDF2-regulated receptors
Transcriptomics to identify gene expression changes resulting from altered RHBDF2 activity
Such integrated approaches provide a systems-level understanding of RHBDF2 function in normal physiology and disease contexts.
Investigation of RHBDF2 post-translational modifications (PTMs) represents a frontier in understanding its functional regulation. Several methodological approaches show particular promise:
Phospho-specific antibodies: Development of antibodies specifically recognizing phosphorylated RHBDF2 at key regulatory sites
Mass spectrometry-based PTM mapping: Using enrichment strategies (TiO₂ for phosphopeptides, lectin affinity for glycopeptides) coupled with MS/MS analysis
Proximity labeling: BioID or TurboID fusion proteins to identify proteins interacting with RHBDF2 in different modification states
Site-directed mutagenesis: Systematic mutation of putative modification sites to serine/threonine/tyrosine to alanine (for phosphorylation) or asparagine to glutamine (for glycosylation)
Researchers should focus on identifying PTMs that regulate:
RHBDF2 trafficking between cellular compartments
Interaction with client proteins like ADAM17
Substrate selectivity and enzyme activation potential
Protein stability and turnover rates
These approaches can reveal how RHBDF2 activity is regulated in different cellular contexts and disease states.
Multiplexed detection of RHBDF2 and its binding partners requires sophisticated imaging and biochemical approaches:
Multiplexed immunofluorescence:
Sequential antibody labeling with fluorophore-conjugated secondary antibodies
Spectral unmixing to resolve overlapping fluorescence signatures
Signal amplification systems for low-abundance targets
Proximity ligation assays (PLA):
Pair RHBDF2 antibodies with antibodies against suspected interaction partners
Use species-specific secondary antibodies with attached oligonucleotides
Ligation and rolling circle amplification to generate fluorescent spots at sites of protein-protein interaction
Co-immunoprecipitation coupled with mass spectrometry:
Use HRP-conjugated RHBDF2 antibodies for pull-down experiments
Label-free quantification or isotope labeling to compare interaction partners under different conditions
Validation of key interactions with targeted Western blotting
These approaches enable comprehensive mapping of the RHBDF2 interactome across different cellular contexts and disease models.