REEP5 antibodies are immunological reagents designed to target the REEP5 protein, a member of the DP1/Yop1p family involved in shaping ER and sarcoplasmic reticulum (SR) membranes . REEP5 is critical for maintaining cardiac SR-tubule junctions and calcium handling , and its dysregulation is linked to diseases such as esophageal squamous cell carcinoma (ESCC) and myocardial infarction . Antibodies against REEP5 are widely used in Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and immunoprecipitation (IP) .
REEP5 antibodies are pivotal in diverse research contexts:
Localization: REEP5 is enriched at SR membranes in cardiomyocytes, as shown by colocalization with RyR2 (ryanodine receptor 2) .
Functional Studies: Knockout models revealed REEP5’s role in maintaining SR-tubule junctions; REEP5 ablation reduces junction length by 40%, impairing calcium release and contractility .
ESCC Prognosis: High REEP5 expression correlates with poor survival in ESCC patients (HR = 1.8, P < 0.05) .
Immune Modulation: REEP5 positively associates with inhibitory checkpoints (CTLA-4, TIM-3) and CD8+ T-cell infiltration, suggesting a role in tumor immunosuppression .
Myocardial Infarction: REEP5 overexpression reduces ER stress markers (PERK, IRE1α) and apoptosis (TUNEL+ cells ↓50%) in murine models .
REEP5 antibodies have advanced understanding of:
Cardiovascular Disease: REEP5’s role in SR integrity highlights therapeutic targets for heart failure .
Oncology: REEP5’s association with immune checkpoints suggests utility in predicting immunotherapy resistance .
ER Stress Disorders: Modulating REEP5 could mitigate ER stress in conditions like diabetes or neurodegeneration .
REEP5 is a membrane protein belonging to the DP1/Yop1p protein family that regulates endoplasmic reticulum (ER) structure and function. It plays a critical role in cardiac health through several mechanisms:
Acts as a regulator of sarco-endoplasmic reticulum (SR/ER) affecting cardiac functions
Significantly enriched in both fetal and adult heart tissue despite broad expression across multiple tissues
Downregulated in myocardial infarction and certain types of cardiomyopathy
Mediates the function of CLEC5A to relieve myocardial infarction by inhibiting ER stress-induced apoptosis
REEP5 depletion can cause SR/ER membrane destabilization and luminal vacuolization, leading to decreased myocyte contractility and disrupted calcium handling, making it a promising target for cardiovascular disease intervention .
Verification of REEP5 antibody specificity is essential for ensuring reliable experimental results:
Blocking experiments: Incubate your antibody with purified REEP5 protein (e.g., 10 μg of bacterially expressed, purified 6xHis-tagged REEP5) before immunostaining to confirm signal elimination
REEP5 knockout controls: Use REEP5 knockout models (available through CRISPR/Cas9-based genome editing) as negative controls to confirm absence of signal
Multiple antibody comparison: Use different REEP5 antibodies targeting distinct epitopes to confirm consistent localization patterns
Western blot analysis: Verify the expected molecular weight (observed at 18-21 kDa, calculated at 21 kDa) and oligomerization patterns (monomers at 17 kDa, dimers at 34 kDa, and trimers at 51 kDa)
Quantitative PCR validation: Confirm REEP5 mRNA levels correlate with protein expression patterns detected by antibody
Proper sample preparation is critical for successful REEP5 detection in cardiac tissue:
For Western Blot Analysis:
Use solubilization buffer containing 6M urea for optimal REEP5 extraction from membrane fractions
Process samples fresh or flash-freeze immediately to prevent protein degradation
Include protease inhibitors to prevent degradation of REEP5 protein
Denature samples at 95°C for 5 minutes in SDS-loading buffer
For Immunohistochemistry:
Fix cardiac tissues in methanol at room temperature for 5 minutes
Block with 10% normal goat serum in PBS for 1 hour at room temperature
For paraffin-embedded sections, perform antigen retrieval with TE buffer pH 9.0 or alternatively with citrate buffer pH 6.0
For Immunofluorescence of Cardiomyocytes:
Optimization of REEP5 antibody concentrations depends on the specific application and antibody used:
Western Blot:
Start with dilutions between 1:2000-1:10000 for polyclonal antibodies
For monoclonal antibodies, higher dilutions (1:20000-1:100000) may be effective
Test a dilution series to determine optimal signal-to-noise ratio
Include positive controls (heart tissue) and negative controls (samples with REEP5 knockdown)
Immunohistochemistry:
Use 1:5000-1:20000 for high-sensitivity monoclonal antibodies
Perform titration experiments to optimize staining
Include both positive control tissues (heart) and negative control tissues (tissues with low REEP5 expression)
Immunofluorescence:
Always include a secondary antibody-only control to assess background
Co-localization studies require careful planning to avoid antibody cross-reactivity and signal interference:
Antibody selection considerations:
Choose REEP5 antibodies from different host species than your other target proteins
When using antibodies from the same host, use directly conjugated primary antibodies
Verify that secondary antibodies do not cross-react
Recommended protocol:
Fix cardiomyocytes with methanol (5 minutes at room temperature)
Perform sequential staining with REEP5 antibody (1:50) and other target antibodies (e.g., RyR2 at 1:50)
Use species-specific secondary antibodies conjugated to spectrally distinct fluorophores (Alexa Fluor 488 and 555)
Include single-staining controls to assess bleed-through
Image using confocal microscopy for optimal resolution of subcellular structures
Analysis approaches:
Calculate Pearson's or Mander's coefficients for quantitative co-localization assessment
Perform line scan analysis across structures of interest
Use deconvolution to improve resolution in densely packed structures
Researchers may encounter several challenges when working with REEP5 antibodies:
Accurate quantification of REEP5 expression changes requires multiple complementary approaches:
Western blot quantification:
Immunohistochemistry quantification:
Standardize image acquisition parameters
Take multiple random fields per section
Use automated image analysis software to quantify staining intensity and distribution
Perform blinded analysis to avoid bias
qPCR for mRNA validation:
Considerations for disease models:
REEP5 shows tissue-specific expression changes in different cardiovascular diseases
In myocardial infarction, REEP5 is downregulated in the infarct penumbra area
In pressure overload-induced heart failure, REEP5 expression is dynamic, showing initial decrease followed by later increase
In idiopathic cardiomyopathy, REEP5 levels may be elevated, while they are decreased in ischemic cardiomyopathy
Differentiating specific from non-specific signals requires appropriate controls:
Essential controls for validating specificity:
Blocking peptide control: Pre-incubate antibody with purified REEP5 protein to abolish specific signal
REEP5 knockdown/knockout: Use CRISPR/Cas9-edited cells or tissues with REEP5 depletion
Antibody isotype control: Use matched isotype control antibody at the same concentration
Secondary antibody only: Omit primary antibody to assess secondary antibody background
Validation approaches:
Compare staining patterns using multiple REEP5 antibodies targeting different epitopes
Verify subcellular localization aligns with REEP5's known function as an ER membrane protein
Confirm molecular weight in Western blot matches predicted size (18-21 kDa)
Correlate protein detection with mRNA expression data
REEP5 antibodies enable sophisticated investigations into ER stress pathways:
Subcellular co-localization studies:
Biochemical interaction studies:
Perform co-immunoprecipitation with REEP5 antibodies to identify novel binding partners
Use REEP5 antibodies in chromatin immunoprecipitation to study transcriptional regulation
Combine with crosslinking approaches to capture transient interactions during ER stress response
Functional studies in disease models:
Monitor REEP5 expression changes during progression of heart disease using Western blot and IHC
Correlate REEP5 levels with markers of ER stress (GRP78, phosphorylated PERK and IRE1α)
Assess impact of REEP5 manipulation on apoptotic markers (cleaved caspase-12, Chop)
Examine REEP5's role in calcium handling using simultaneous calcium imaging and immunofluorescence
Resolving conflicting reports requires careful experimental design:
Standardized disease model characterization:
Temporal profiling approaches:
Regional analysis within cardiac tissue:
Examine region-specific differences (infarct core vs. penumbra vs. remote areas)
Use laser capture microdissection to isolate specific regions
Perform detailed immunohistochemical mapping across tissue sections
Multi-level analysis:
REEP5 antibodies can be integrated into advanced single-cell approaches:
Single-cell protein profiling:
Use flow cytometry with REEP5 antibodies to characterize cellular heterogeneity in heart tissue
Apply Imaging Mass Cytometry (IMC) with metal-conjugated REEP5 antibodies for multiparameter analysis
Implement CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) to correlate REEP5 protein levels with transcriptome
Spatial transcriptomics integration:
Combine REEP5 immunohistochemistry with spatial transcriptomics to map protein-mRNA relationships
Use multiplexed immunofluorescence to study REEP5 in relation to cell-type specific markers
Apply nearest-neighbor analysis to identify spatial relationships between REEP5-expressing cells and other cardiac cell populations
Live cell imaging approaches:
Use cell-permeable, fluorescently-labeled REEP5 antibody fragments to track dynamic changes
Implement FRET-based biosensors to monitor REEP5 interactions with binding partners
Apply super-resolution microscopy techniques (STORM, PALM) to resolve REEP5 nano-organization within ER membranes
Cancer research applications:
Examine REEP5's role in T-cell infiltration in esophageal squamous cell carcinoma using immunoprofiling
Investigate correlations between REEP5 and immune checkpoints (CTLA-4, TIM-3, HVEM) via multiplex IHC
Use single-cell clustering analysis to identify cell-specific expression patterns of REEP5 between cancerous and adjacent tissues
Several emerging technologies could significantly advance REEP5 research:
Nanobody and recombinant antibody technologies:
Development of REEP5-specific nanobodies with improved tissue penetration
Creation of bispecific antibodies targeting REEP5 and binding partners simultaneously
Engineering of intrabodies for live-cell visualization of REEP5 dynamics
Advanced imaging approaches:
Implementation of expansion microscopy to visualize REEP5 within complex ER networks
Application of correlative light and electron microscopy to study REEP5 at ultrastructural level
Development of light-sheet microscopy protocols for 3D visualization of REEP5 in whole-heart preparations
Controlled protein modulation:
REEP5-targeting protein degradation systems (PROTAC, dTAG)
Optogenetic control of REEP5 function using antibody-guided photosensitizers
Antibody-drug conjugates for targeted delivery to REEP5-expressing tissues
REEP5 antibodies could facilitate therapeutic development through several approaches:
Target validation strategies:
Use antibodies to map REEP5 expression in patient samples
Correlate REEP5 levels with disease severity and treatment response
Identify patient subgroups that might benefit from REEP5-targeted therapies
Therapeutic screening platforms:
Develop high-content screening assays using REEP5 antibodies to identify compounds that modulate REEP5 expression or localization
Create reporter systems to monitor ER stress responses in relation to REEP5 function
Screen for biologics that stabilize REEP5-mediated ER membrane organization
Potential therapeutic mechanisms:
Biomarker development:
Explore REEP5 as a potential biomarker for cardiovascular disease progression
Develop highly sensitive immunoassays for detecting REEP5 in circulation
Investigate REEP5 post-translational modifications as disease-specific indicators