RTP3 (Receptor-transporting protein 3), also known as TMEM7 or LTM1, is a single-pass type III membrane protein encoded by the RTP3 gene located on human chromosome 3p21.31 . It belongs to the TMEM7 family and facilitates the functional expression of bitter taste receptors . In oncology, RTP3 is implicated in hepatocellular carcinoma (HCC) progression via the STAT3 signaling pathway .
The RTP3 antibody is a polyclonal or monoclonal reagent designed to detect RTP3 in research applications. It is widely used in studies exploring cancer biology, taste perception, and protein trafficking.
Recent studies highlight RTP3’s role in HCC pathogenesis:
TRPM8-RTP3-STAT3 Axis: TRPM8 (a cold-sensing ion channel) overexpression suppresses HCC proliferation and metastasis by downregulating RTP3 and inhibiting STAT3 activation .
AD80 Therapeutic Potential: The multikinase inhibitor AD80 upregulates TRPM8, indirectly repressing RTP3 and STAT3, thereby inhibiting HCC growth in vitro and in vivo .
Prognostic Marker: Low TRPM8/RTP3 expression correlates with advanced HCC histological grades and poor patient survival .
Therapeutic Target: Targeting the TRPM8-RTP3-STAT3 pathway may offer novel treatment strategies .
Western Blot (WB): Detects RTP3 at ~27 kDa in human liver, testis, and kidney lysates .
Immunohistochemistry (IHC): Localizes RTP3 in circumvallate papillae and testis tissues .
Functional Studies: Validates RTP3’s role in bitter taste receptor trafficking and cancer signaling .
RTP3 (Receptor Transporter Protein 3) is a membrane-localized protein with a length of 232 amino acid residues and a molecular weight of approximately 27 kDa in humans . It belongs to the TMEM7 protein family and plays a crucial role in promoting functional cell surface expression of bitter taste receptors, specifically TAS2R16 and TAS2R43 . The significance of RTP3 in research stems from its high expression in adult liver tissue and its conserved presence across several mammalian species including mouse, rat, bovine, and chimpanzee . Understanding RTP3's function has implications for taste perception research, membrane protein trafficking, and potentially liver-specific cellular functions.
RTP3 antibodies target various epitope regions of the protein, with commercially available options covering different amino acid sequences including AA 1-211, AA 21-120, and AA 81-180 . The choice of epitope significantly impacts experimental outcomes because:
Accessibility: Membrane-embedded regions may be less accessible than extracellular domains
Conservation: Epitopes with higher evolutionary conservation allow cross-species applications
Post-translational modifications: Modifications near certain epitopes may block antibody binding
Functional domains: Antibodies targeting functional domains may inhibit protein activity
For optimal results in functional studies, researchers should select antibodies targeting the AA 21-120 region, which includes important functional domains . For protein expression studies, antibodies against the AA 1-211 region provide broader coverage of the protein . The AA 81-180 region contains the sequence "RGQVKMRVFT QRCKKCPQPL FEDPEFTQEN ISRILKNLVF RILKKCYRGR FQLIEEVPMI KDISLEGPHN SDNCEACLQG FCAGPIQVTS LPPSQTPRVH" which includes several important functional motifs .
The choice of host animal in which anti-RTP3 antibodies are raised significantly influences antibody characteristics and experimental applications. Based on available information:
Rabbit-derived polyclonal RTP3 antibodies demonstrate broad applicability across multiple techniques including Western Blot, ELISA, immunohistochemistry, and flow cytometry . These antibodies typically offer high sensitivity due to their recognition of multiple epitopes on the RTP3 protein.
Mouse-derived polyclonal RTP3 antibodies show more limited application range, primarily validated for Western Blot and ELISA techniques . These antibodies may offer advantages when performing multi-color immunostaining alongside rabbit-derived antibodies against other targets.
The host species selection impacts:
Background staining in tissues of the same species (increased background when using mouse antibodies on mouse tissues)
Secondary antibody compatibility
Recognition of post-translational modifications (which can vary based on the host's immune response)
Cross-reactivity profile (rabbit polyclonals generally show broader cross-reactivity across species)
Optimizing Western blotting protocols for RTP3 antibody detection requires careful consideration of several technical parameters:
Sample preparation:
Use RIPA buffer with protease inhibitors for membrane protein extraction
Heat samples at 70°C (not 95°C) for 10 minutes to prevent membrane protein aggregation
Use fresh samples when possible as RTP3 can degrade during freeze-thaw cycles
Gel electrophoresis:
Use 10-12% polyacrylamide gels to properly resolve the 27 kDa RTP3 protein
Load 20-30 μg of total protein per lane for liver samples (where RTP3 is highly expressed)
Include positive controls from liver tissue extracts
Transfer conditions:
Use PVDF membranes (0.45 μm pore size) for optimal protein binding
Transfer at 100V for 1 hour in 10% methanol-containing transfer buffer
Verify transfer efficiency with Ponceau S staining
Antibody incubation:
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Dilute primary RTP3 antibodies 1:500 to 1:1000 depending on specific product
Incubate with primary antibody overnight at 4°C
Use appropriate species-specific HRP-conjugated secondary antibodies at 1:5000 dilution
Detection:
Use enhanced chemiluminescence (ECL) detection reagents
Expect a band at approximately 27 kDa for human RTP3
This methodology has been validated with both rabbit-derived and mouse-derived anti-RTP3 antibodies across multiple studies .
Ensuring antibody specificity is crucial for generating reliable research data. For RTP3 antibodies, implement these validation steps:
Positive and negative controls:
Peptide competition assay:
Pre-incubate the antibody with excess immunizing peptide (e.g., the specific AA sequence used to generate the antibody)
Run parallel Western blots or IHC with blocked and unblocked antibody
Specific binding should be eliminated in the peptide-blocked sample
Cross-reactivity assessment:
Test the antibody against recombinant RTP3 and closely related family members
Verify signal reduction in samples with reduced RTP3 expression
Check antibody reactivity in multiple species if cross-species reactivity is claimed
Orthogonal method validation:
Compare results from different antibody clones targeting distinct RTP3 epitopes
Validate findings using nucleic acid-based methods (RT-PCR, RNA-seq)
Consider using tagged RTP3 constructs for expression with subsequent detection by tag-specific antibodies
Mass spectrometry confirmation:
Immunoprecipitate RTP3 using the antibody
Confirm the identity of the precipitated protein by mass spectrometry
This provides definitive evidence of antibody specificity
These validation steps significantly enhance confidence in experimental results and minimize the risk of antibody-based artifacts .
Optimizing immunohistochemical (IHC) detection of RTP3 in tissue sections requires attention to several critical parameters:
Tissue preparation:
Fix tissues in 10% neutral-buffered formalin for 24 hours
Process and embed in paraffin following standard protocols
Cut sections at 4-5 μm thickness
Antigen retrieval (critical for RTP3 detection):
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes
Alternative: EDTA buffer (pH 9.0) if citrate buffer yields insufficient signal
Allow slides to cool slowly to room temperature
Blocking and antibody incubation:
Block endogenous peroxidase with 3% hydrogen peroxide for 10 minutes
Apply protein block (5% normal serum) for 30 minutes
Incubate with primary anti-RTP3 antibody at 1:100-1:200 dilution overnight at 4°C
Use antibodies validated for IHC-p (paraffin sections), particularly those targeting the AA 21-120 region
Detection system:
Use biotin-free polymer detection systems to minimize background
Develop with DAB chromogen for 5-10 minutes (monitor under microscope)
Counterstain with hematoxylin for 1-2 minutes
Controls and interpretation:
Include liver tissue as positive control
Use isotype control antibodies as negative controls
Expect membranous staining pattern in positive cells
Evaluate both staining intensity and percentage of positive cells
This protocol has been optimized based on the properties of available RTP3 antibodies and the membrane localization of the target protein .
Co-immunoprecipitation (Co-IP) with RTP3 antibodies provides valuable insights into protein-protein interactions. The following methodological approach is recommended:
Cell/tissue preparation:
Harvest cells or tissues expressing RTP3 (liver tissue or appropriate cell lines)
Lyse in gentle lysis buffer (1% NP-40, 150 mM NaCl, 50 mM Tris pH 7.4) with protease inhibitors
Clear lysates by centrifugation at 14,000 g for 15 minutes at 4°C
Antibody selection and coupling:
Immunoprecipitation:
Incubate cleared lysate with antibody-coupled beads overnight at 4°C with gentle rotation
Wash beads 4-5 times with cold lysis buffer containing reduced detergent (0.1% NP-40)
Elute bound proteins with either gentle elution buffer or by boiling in SDS sample buffer
Analysis of interacting proteins:
Validation of interactions:
Confirm interactions by reverse Co-IP using antibodies against identified partners
Validate with in vitro binding assays using recombinant proteins
Consider proximity ligation assays to confirm interactions in intact cells
This approach enables researchers to map the RTP3 interactome, particularly focusing on its role in trafficking bitter taste receptors to the cell surface .
Optimizing flow cytometry for RTP3 detection requires specialized procedures due to its membrane localization:
Sample preparation:
Harvest cells gently using non-enzymatic cell dissociation solution
Fix with 2% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1% saponin (not Triton X-100) to preserve membrane integrity
Antibody selection and staining:
Use fluorochrome-conjugated anti-RTP3 antibodies when available (such as FITC-conjugated)
Alternatively, use primary anti-RTP3 antibodies validated for flow cytometry followed by fluorophore-conjugated secondary antibodies
Antibodies targeting the AA 21-120 region show optimal results for flow cytometry
Incubate with antibody in buffer containing 0.1% saponin for 45-60 minutes at 4°C
Protocol optimization:
Titrate antibody concentration to determine optimal signal-to-noise ratio
Include appropriate isotype controls matched to primary antibody
Perform compensation controls when multiplexing with other markers
Use viability dye to exclude dead cells from analysis
Data acquisition and analysis:
Collect at least 10,000 events per sample
Gate on single cells using FSC-A vs. FSC-H
Analyze RTP3 expression relative to known markers of target cell populations
Consider co-staining for bitter taste receptors to analyze co-expression patterns
Validation strategies:
Include positive controls (liver-derived cells) and negative controls
Compare results with Western blot quantification
Verify specificity with competitive blocking using immunizing peptide
This approach allows quantitative assessment of RTP3 expression across different cell types and experimental conditions .
Investigating RTP3 subcellular localization and trafficking requires multi-modal approaches:
Immunofluorescence microscopy:
Fix cells with 4% paraformaldehyde for 15 minutes
Permeabilize with 0.1% saponin to preserve membrane structures
Use anti-RTP3 antibodies at 1:100-1:200 dilution
Co-stain with markers for specific cellular compartments:
Plasma membrane (Na+/K+-ATPase)
Endoplasmic reticulum (calnexin)
Golgi apparatus (GM130)
Endosomes (Rab5, Rab7)
Analyze using confocal microscopy for precise co-localization assessment
Live-cell imaging with fluorescent protein fusions:
Create RTP3-GFP or RTP3-mCherry fusion constructs
Transfect cells with minimal expression to avoid overexpression artifacts
Perform time-lapse imaging to track RTP3 trafficking
Use photoactivatable or photoconvertible tags for pulse-chase analysis
Biochemical fractionation:
Separate cellular compartments using differential centrifugation
Prepare membrane, cytosolic, and nuclear fractions
Analyze RTP3 distribution by Western blotting using specific anti-RTP3 antibodies
Include markers for each fraction to confirm separation quality
Proximity labeling approaches:
Create BioID or APEX2 fusions with RTP3
Identify proximal proteins in different cellular compartments
Map the spatial organization of RTP3 in the cell
Super-resolution microscopy:
Apply STORM or PALM imaging using appropriately labeled secondary antibodies
Achieve nanometer-scale resolution of RTP3 distribution
Quantify clustering and co-localization with interacting proteins
These methodologies provide complementary data on RTP3's dynamic localization, particularly in relation to its role in trafficking bitter taste receptors to the cell surface .
Understanding potential artifacts in RTP3 antibody-based experiments is critical for accurate data interpretation:
Common causes of false positive results:
Cross-reactivity with structurally similar proteins, particularly other TMEM family members
Non-specific binding to highly expressed liver proteins
Inappropriate secondary antibody selection causing background signal
Insufficient blocking leading to hydrophobic interactions with membrane fractions
Over-development of immunohistochemistry or Western blot signals
Common causes of false negative results:
Epitope masking due to protein-protein interactions or post-translational modifications
Inadequate sample preparation failing to expose membrane-embedded epitopes
Insufficient antigen retrieval in fixed tissues
Protein degradation during sample preparation
Sub-optimal antibody concentration or incubation conditions
Mitigation strategies:
Always include positive and negative controls
Validate results with at least two antibodies targeting different epitopes
Perform peptide competition assays to confirm specificity
Use genetic approaches (knockdown/knockout) to validate antibody specificity
Consider the native expression level of RTP3 in your experimental system
Technical considerations:
For Western blot: ensure complete protein transfer and appropriate blocking
For IHC: optimize antigen retrieval and minimize background with appropriate blockers
For flow cytometry: careful gating and compensation to avoid autofluorescence artifacts
These considerations help researchers distinguish genuine RTP3 signals from technical artifacts .
When faced with discrepancies between different antibody-based detection methods for RTP3, employ this systematic approach to interpretation:
Method-specific considerations:
Western blot detects denatured protein and reveals molecular weight information
IHC preserves tissue architecture but may have reduced epitope accessibility
Flow cytometry provides quantitative single-cell data but requires cell dissociation
IP methods depend on native protein conformation and accessibility in solution
Epitope-dependent factors:
Different epitopes may be differentially accessible in various applications
Antibodies targeting AA 21-120 perform better in IHC and flow cytometry applications
Antibodies targeting AA 1-211 may be superior for Western blot applications
Post-translational modifications may block specific epitopes in a context-dependent manner
Hierarchical resolution approach:
Determine which method provides higher specificity through controls
Consider orthogonal methods (mRNA analysis, tagged protein expression)
Evaluate which method aligns with known biology of RTP3
Assess technical quality and reproducibility of each method
Integration strategy:
Create a consensus view based on multiple methods
Weight results based on validated controls for each method
Document methodological variables that may explain discrepancies
Consider biological variables (splice variants, protein modifications)
This approach enables researchers to resolve apparent contradictions and develop a more accurate understanding of RTP3 biology across experimental systems .
Selecting appropriate quantification methods for RTP3 antibody signals is critical for generating reliable and reproducible data:
Western blot quantification:
Use densitometry with background subtraction
Normalize to appropriate loading controls (β-actin, GAPDH)
For membrane proteins like RTP3, consider normalizing to Na+/K+-ATPase or cadherin
Apply linear range detection methods (avoid saturated signals)
Present data as fold change relative to controls
Immunohistochemistry quantification:
Use H-score system (intensity × percentage of positive cells)
Alternative: Automated image analysis with tissue segmentation
Score membrane staining separately from cytoplasmic signals
Blind scoring by multiple observers to reduce bias
Include scale bars and representative images at multiple magnifications
Flow cytometry quantification:
Report median fluorescence intensity (MFI) rather than mean
Calculate signal-to-noise ratio relative to isotype controls
Use molecules of equivalent soluble fluorochrome (MESF) for absolute quantification
Apply proper compensation when multiplexing
Present percentage of positive cells based on objective gating strategies
Immunoprecipitation quantification:
Normalize to input material
Compare to IgG control pull-downs
Consider semi-quantitative ranking of interaction strengths
Include loading controls for input samples
Statistical analysis:
Apply appropriate statistical tests based on data distribution
Report biological and technical replicates separately
Consider power analysis to determine adequate sample sizes
Use non-parametric tests when appropriate for IHC scoring
These quantification strategies enhance the rigor and reproducibility of RTP3 antibody-based research .
The decision between monoclonal and polyclonal RTP3 antibodies should be guided by application-specific requirements:
Polyclonal RTP3 antibodies advantages:
Higher sensitivity due to recognition of multiple epitopes
Greater tolerance to minor protein denaturation or modifications
Better performance in applications requiring high detection sensitivity
Currently more widely validated across multiple applications
Available from rabbit and mouse host species with various conjugations
Monoclonal RTP3 antibodies advantages:
Superior reproducibility between experiments and lots
Higher specificity for particular epitopes
Reduced batch-to-batch variation
Better performance in quantitative applications requiring precise standardization
Lower background in some applications
Application-specific recommendations:
For Western blot: Both types work well, with polyclonals providing higher sensitivity
For IHC: Polyclonal antibodies often provide stronger signal in paraffin sections
For flow cytometry: Monoclonals may provide more consistent results
For quantitative studies: Monoclonals offer better standardization
For detecting modified forms: Polyclonals that recognize multiple epitopes
Selection matrix for RTP3 antibodies:
Application | Preferred Type | Recommended Epitope | Key Consideration |
---|---|---|---|
Western Blot | Either | AA 81-180 | Signal specificity |
IHC-p | Polyclonal | AA 21-120 | Antigen retrieval |
Flow Cytometry | Either | AA 21-120 | Membrane integrity |
IP | Polyclonal | AA 1-211 | Native conformation |
ELISA | Either | Any validated | Standardization |
This guidance helps researchers select the most appropriate antibody format for their specific experimental goals .
The epitope-targeting strategy significantly influences RTP3 antibody performance across experimental systems:
N-terminal region antibodies (AA 1-211):
Middle region antibodies (AA 21-120):
Internal region antibodies (AA 81-180):
Experimental system considerations:
Cell lines: Middle region antibodies perform well in most cultured cell systems
Tissues: Antibodies targeting AA 21-120 show optimal IHC performance in FFPE tissues
Recombinant systems: N-terminal antibodies useful for tagged-protein detection
Species cross-reactivity: Internal epitopes show higher conservation across species
Functional domain targeting:
Zinc finger domains: Use AA 81-180 antibodies to study DNA binding functions
Membrane-associated regions: AA 21-120 antibodies for trafficking studies
Protein-protein interaction domains: Consider the impact of antibody binding on complex formation
This knowledge enables researchers to strategically select antibodies based on their specific experimental questions and systems .
Implementing robust controls is essential for generating reliable data with RTP3 antibodies in functional studies:
Antibody specificity controls:
Peptide competition/blocking: Pre-incubate antibody with immunizing peptide
Genetic validation: Use RTP3 knockout/knockdown samples
Recombinant protein: Test antibody against purified RTP3 protein
Overexpression: Compare endogenous signal with overexpressed RTP3
Technical controls:
Functional study-specific controls:
For trafficking studies:
For protein-protein interaction studies:
Perform reciprocal co-immunoprecipitations
Include non-interacting protein controls
Verify interactions with multiple antibodies targeting different epitopes
Quantitative controls:
Standard curves with recombinant protein (for quantitative applications)
Internal reference samples across experiments for normalization
Biological replicates from independent sources
Technical replicates to assess method reproducibility
Documentation requirements:
Record complete antibody information (supplier, catalog number, lot number)
Document detailed experimental conditions
Maintain original unprocessed data alongside analyzed results
Report all controls in publications and presentations
The field of RTP3 antibody applications continues to evolve with several emerging areas of research significance:
Single-cell analysis applications:
Integration of RTP3 antibodies in mass cytometry (CyTOF) panels for deep phenotyping
Single-cell Western blotting to examine RTP3 expression heterogeneity
Spatial proteomics using antibody-based detection methods to map RTP3 distribution within tissue microenvironments
Combined protein-transcript analysis at single-cell resolution using antibodies with in situ hybridization
High-throughput screening applications:
Development of RTP3 antibody-based biosensors for real-time trafficking studies
Incorporation into automated immunoassay platforms for biomarker quantification
Application in cell-based screens to identify modulators of bitter taste receptor trafficking
Advanced imaging techniques:
Super-resolution microscopy with specialized secondary antibody conjugates
Expansion microscopy for nanoscale visualization of RTP3 distribution
Cryo-electron tomography using antibody-gold labeling for 3D ultrastructural analysis
Intravital imaging using minimally invasive antibody-based probes
Therapeutic and diagnostic development:
Internalization studies to assess RTP3 as a potential drug delivery target
Development of conformation-specific antibodies to distinguish functional states
Exploration of RTP3's role in liver pathophysiology using specific antibodies
These emerging applications represent cutting-edge directions in RTP3 research, building upon fundamental knowledge of its structure and function while leveraging technological advances in antibody-based detection methods .
When preparing RTP3 antibody data for publication, researchers should follow these comprehensive validation and reporting guidelines:
Essential validation experiments to perform:
Specificity testing using positive and negative controls
Demonstration of expected molecular weight by Western blot
Peptide competition assays showing signal reduction
Correlation with mRNA expression or orthogonal protein detection methods
Application-specific controls (as detailed in section 5.3)
Complete antibody documentation:
Commercial source, catalog number, and lot number
Clone identifier for monoclonals or immunogen details for polyclonals
Host species and antibody isotype
Exact epitope or immunogen sequence
RRID (Research Resource Identifier) when available
Detailed methodology reporting:
Antibody concentration/dilution used
Incubation conditions (time, temperature, buffer composition)
Detection methods (secondary antibodies, detection chemistry)
Image acquisition parameters (exposure, gain settings)
Quantification methods with statistical approaches
Transparent data presentation:
Include full unprocessed blot images with molecular weight markers
Show representative images alongside quantification
Present both positive and negative results
Include all relevant controls in figures or supplements
Declare any antibody validation limitations
Adherence to community guidelines:
Follow journal-specific antibody reporting requirements
Consider the International Working Group for Antibody Validation (IWGAV) guidelines
Address the "Five Pillars" of antibody validation when applicable