Key uses in experimental workflows:
ELISA: Quantifying RUBCN expression levels in serum or cell lysates .
Western blot: Detecting RUBCN at ~108.6 kDa in human tissues (e.g., liver, spleen) .
Autophagy studies: Investigating RUBCN’s role in modulating PI3K complex II activity and autophagosome maturation .
Example findings using RUBCN-targeting tools:
RUBCN-deficient mice exhibit hypersensitivity to acute kidney injury (AKI), partially rescued by co-deletion of MLKL .
RUBCN regulates lipid metabolism and autolysosome formation, with opposing effects to its paralog RUBCNL/Pacer .
Selected validation metrics from comparable antibodies:
Proteintech’s RUBCN antibody (21444-1-AP): Validated in WB (mouse liver, HeLa cells), IHC (human testis), and IF/ICC .
Assay Genie’s CAB13169: Detects RUBCN at 1:500–1:2000 dilution in WB .
Cross-reactivity notes:
Most commercial RUBCN antibodies show reactivity with human, mouse, and rat samples .
No significant cross-reactivity with RUBCNL/Pacer reported .
Notable suppliers of RUBCN antibodies (HRP and unconjugated):
| Supplier | Conjugate | Price (100 µL) | Applications |
|---|---|---|---|
| CUSABIO Technology LLC | HRP | $299 | ELISA |
| Fine Biotech Co., Ltd. | Unconjugated | $240 | WB, ELISA, IHC |
| Abbexa Ltd | Unconjugated | $261 | WB, ICC, IF, IHC |
| Cell Signaling Technology | Unconjugated | $327 | WB (validated in 57 studies) |
HRP-conjugated versions are less common than unconjugated variants, necessitating careful supplier selection .
RUBCN’s functional roles:
Modulates necroptosis and inflammation in kidney injury models .
Implicated in chronic kidney disease (CKD) with age-dependent phenotypes .
Therapeutic research implications:
RUBCN-deficient models help study autophagy-related diseases (e.g., neurodegeneration, cancer) .
Antibodies enable tracking RUBCN expression changes under stress or drug treatment .
Buffer compatibility: Avoid amine-containing buffers (e.g., Tris) during conjugation, as they interfere with HRP labeling .
Detection: Use high-sensitivity substrates (e.g., enhanced chemiluminescence for WB, TMB for ELISA) .
Controls: Include RUBCN knockout lysates or siRNA-treated samples to confirm specificity .
RUBCN (RUN domain and cysteine-rich domain containing, Beclin 1-interacting protein), also known as Rubicon, functions as a negative regulator of autophagy by inhibiting the fusion of autophagosomes and lysosomes . RUBCN is particularly significant in aging research as its expression increases in aged tissues across multiple model organisms including worms, flies, and mice at both transcript and protein levels . The protein plays a crucial role in cellular homeostasis by regulating autophagic flux, which has implications for various pathological conditions including metabolic disorders and kidney function . RUBCN's inhibitory effect on autophagy makes it a valuable target for studying cellular degradation pathways, stress responses, and age-related pathologies.
HRP (horseradish peroxidase) conjugation provides a highly sensitive detection method for antibodies by catalyzing a chemiluminescent reaction. When conjugated to RUBCN antibodies, HRP enables precise visualization of the target protein in techniques such as Western blotting, immunohistochemistry, and ELISA. The conjugation process attaches HRP molecules to the antibody through chemical crosslinking while preserving the antibody's binding affinity . This direct conjugation eliminates the need for secondary antibodies, reducing background noise and increasing detection specificity. The enzymatic activity of HRP creates an amplification effect, allowing researchers to detect even low abundance proteins like RUBCN in complex biological samples .
RUBCN expression exhibits significant tissue-specific variation and is dynamically regulated during aging and disease states. Studies across model organisms have demonstrated that RUBCN expression increases in aged tissues of worms, flies, and mice . In mice specifically, tissue-specific knockout models have revealed distinct phenotypes:
When designing experiments to study RUBCN, researchers should consider these tissue-specific variations and select appropriate controls matching the experimental context to ensure accurate interpretation of results.
For optimal Western blot detection of RUBCN using HRP-conjugated antibodies, follow these evidence-based methodological guidelines:
Sample preparation: Extract proteins using RIPA buffer supplemented with protease inhibitors. For tissues with high lipid content (where RUBCN is often enriched), consider specialized extraction buffers.
Gel electrophoresis: Use 8-10% polyacrylamide gels as RUBCN is approximately 130 kDa. Load 20-40 μg of total protein per well, depending on RUBCN abundance in your sample.
Transfer conditions: Transfer proteins to nitrocellulose membrane at 100V for 60-90 minutes in cold transfer buffer containing 20% methanol .
Blocking: Block membranes with 5% non-fat dry milk in TBST for 1 hour at room temperature to minimize non-specific binding.
Primary antibody incubation: When using HRP-conjugated RUBCN antibodies, dilute according to manufacturer recommendations (typically 1:1000 to 1:5000) in blocking solution and incubate overnight at 4°C.
Wash steps: Perform 4-5 washes with TBST, 5 minutes each, to remove unbound antibody.
Detection: Use enhanced chemiluminescent substrates optimized for HRP detection such as Azure Radiance chemiluminescent substrates . Expose to appropriate detection system based on expected signal strength.
Stripping and reprobing: If needed, use commercial HRP stripping buffer before reprobing with additional antibodies.
This protocol provides superior detection sensitivity while minimizing background, crucial for accurate quantification of RUBCN, especially in comparative studies across different physiological conditions.
Rigorous validation of RUBCN antibodies is essential for generating reliable research data. Implement these comprehensive validation steps:
Positive and negative controls: Use tissue samples from RUBCN knockout models as negative controls . PTEC-specific RUBCN-deficient mice tissues provide excellent negative controls for kidney research, while tissues with known high RUBCN expression (aged tissues) serve as positive controls .
Peptide competition assay: Pre-incubate the antibody with purified RUBCN peptide before application to demonstrate binding specificity.
Cross-reactivity assessment: Test the antibody on samples from multiple species if conducting comparative studies. RUBCN is conserved across worms (rub-1), flies, and mammals, but antibodies may show species-specific detection patterns .
Molecular weight verification: Confirm that the detected band appears at the expected molecular weight (approximately 130 kDa for full-length human RUBCN).
siRNA/shRNA knockdown validation: In cell culture systems, compare RUBCN detection in control versus RUBCN-knockdown cells to validate signal specificity.
Immunoprecipitation followed by mass spectrometry: For ultimate validation, immunoprecipitate the target using the antibody and confirm identity by mass spectrometry.
Co-localization studies: For immunocytochemistry applications, verify that RUBCN antibody staining co-localizes with known RUBCN interaction partners like Beclin-1 or autophagosomal markers.
When encountering challenges with HRP-conjugated RUBCN antibodies, these evidence-based troubleshooting approaches can resolve specific issues:
For challenging tissues like brain or kidney where RUBCN detection can be problematic, consider tissue-specific extraction protocols. For kidney proximal tubular epithelial cells specifically, specialized lysis buffers containing phosphatase inhibitors improve RUBCN detection, as demonstrated in studies with PTEC-specific RUBCN-deficient mice .
RUBCN antibodies serve as powerful tools for investigating autophagy dynamics across various disease models, offering several methodological approaches:
Autophagy flux assessment: By measuring RUBCN levels alongside autophagy markers like LC3-II and p62, researchers can evaluate autophagic flux inhibition. Higher RUBCN expression correlates with decreased autophagosome-lysosome fusion, a hallmark of many diseases .
Genetic interaction studies: In models where autophagy genes are manipulated, RUBCN antibodies help assess compensatory mechanisms. For example, in studies examining the effects of Rubcn deficiency on kidney proximal tubular epithelial cells, RUBCN antibodies confirmed knockout efficiency while simultaneously measuring changes in other autophagy regulators .
Stress response profiling: During cellular stress (nutrient deprivation, oxidative stress), RUBCN antibodies can track dynamic changes in protein levels and post-translational modifications. This approach has revealed that RUBCN serves as a stress-responsive autophagy regulator in multiple disease contexts.
Aging-related autophagy impairment: Since RUBCN expression increases with age across multiple species , RUBCN antibodies provide a valuable biomarker for age-related autophagy decline. Researchers can quantitatively assess how interventions aimed at extending lifespan affect RUBCN levels.
Therapeutic compound screening: When evaluating compounds designed to enhance autophagy, RUBCN antibodies help determine whether therapeutic effects occur through RUBCN modulation or independent pathways.
In metabolic disease models specifically, RUBCN antibodies have helped establish that sustained high autophagic flux resulting from RUBCN deficiency can lead to metabolic syndrome with accelerated mobilization of phospholipids from cellular membranes to lysosomes , demonstrating the complex role of autophagy in metabolic homeostasis.
When applying RUBCN antibodies across different model organisms, researchers must account for these critical considerations:
Sequence homology and epitope conservation: RUBCN is evolutionarily conserved but shows sequence variations across species. For example, the C. elegans homolog (rub-1) and mammalian RUBCN share functional domains but differ in regions that may affect antibody recognition . Select antibodies raised against conserved epitopes when conducting cross-species studies.
Expression pattern differences: RUBCN expression patterns vary considerably between organisms:
In C. elegans, rub-1 expression can be visualized using rub-1::EGFP translational fusion constructs
In mice, RUBCN expression increases with age in multiple tissues, but baseline expression varies by tissue type
In humans, RUBCN expression patterns may differ from model organisms, requiring validation in human samples
Isoform specificity: Different species may express multiple RUBCN isoforms. Ensure your antibody detects the relevant isoform for your research question.
Extraction protocol optimization:
Validation standards: Use genetic models appropriate to each organism:
By addressing these considerations, researchers can ensure robust cross-species comparisons in RUBCN research, particularly valuable for evolutionary studies of autophagy regulation.
The choice of fixation and permeabilization methods significantly impacts RUBCN antibody performance in immunohistochemistry and immunocytochemistry applications. Based on empirical evidence:
Fixation protocols:
Paraformaldehyde (4%): Provides good preservation of RUBCN epitopes while maintaining cellular architecture. Optimal fixation time is 10-15 minutes for cultured cells and 24 hours for tissue sections.
Methanol fixation: Generally suboptimal for RUBCN detection as it can disrupt protein conformation of membrane-associated proteins like RUBCN.
Glutaraldehyde: Should be avoided as it causes excessive crosslinking that masks RUBCN epitopes.
Permeabilization methods:
Triton X-100 (0.1-0.3%): Effective for most applications, but concentration and duration should be optimized. Excessive permeabilization can lead to loss of membrane-associated RUBCN.
Saponin (0.05-0.1%): Preferable for preserving membrane structures where RUBCN localizes.
Digitonin (10-50 μg/ml): Useful for selective permeabilization of plasma membrane while preserving internal membranes, beneficial for distinguishing populations of RUBCN.
Antigen retrieval techniques:
Heat-induced epitope retrieval: Using citrate buffer (pH 6.0) improves detection in formalin-fixed tissues.
Enzymatic retrieval: Generally not recommended as it may degrade membrane structures where RUBCN localizes.
Tissue-specific considerations:
Kidney sections: Require gentler permeabilization (0.1% Triton X-100 for 5 minutes) to preserve PTEC morphology while allowing antibody access .
Brain tissue: Benefits from longer antigen retrieval due to lipid-rich composition.
Liver sections: Often require lipid extraction steps to reduce background.
These protocol variations should be systematically tested when establishing RUBCN immunodetection in new experimental systems to ensure optimal signal-to-noise ratio and biological relevance of the results.
Interpreting changes in RUBCN levels requires nuanced analysis across different experimental contexts:
Age-related increases: Multiple studies demonstrate that RUBCN expression increases with age across species (worms, flies, mice) . This increase correlates with declining autophagy capacity and may contribute to age-related pathologies. When interpreting age-dependent RUBCN changes:
Compare against appropriate age-matched controls
Consider tissue-specific variance in expression patterns
Evaluate concurrent changes in other autophagy regulators
Disease contexts: Elevated RUBCN levels observed in disease states should be interpreted in these frameworks:
Causative factor vs. compensatory response: Determine whether RUBCN upregulation precedes pathology (suggesting causative role) or follows it (suggesting compensatory mechanism)
Tissue-specific effects: RUBCN deficiency produces different outcomes depending on tissue context—improving liver steatosis in hepatocytes while potentially contributing to metabolic syndrome in kidney PTECs
Intervention response: Changes in RUBCN following therapeutic interventions may serve as biomarkers of treatment efficacy
Molecular context: RUBCN data should be interpreted alongside:
Autophagy flux markers (LC3-II, p62/SQSTM1)
Lysosomal function indicators (LAMP1, cathepsins)
Upstream regulators (mTOR signaling components)
Post-translational modifications of RUBCN itself
Quantitative considerations: When performing Western blot analysis:
Use densitometry normalized to appropriate loading controls
Apply statistical analysis appropriate for the distribution of your data
Consider non-linear relationships between RUBCN levels and functional outcomes
The relationship between RUBCN levels and autophagy is not always linear—even modest increases can significantly impact autophagy flux, while complete absence may trigger compensatory mechanisms that complicate interpretation.
Rigorous experimental design requires specific controls when manipulating RUBCN expression:
Genetic manipulation controls:
Conditional knockout models: Use Cre-negative littermates with the same floxed Rubcn allele as controls, as demonstrated in PTEC-specific rubcn-deficient mice studies
Global knockout: Heterozygous littermates often serve as better controls than wild-type, as they share more genetic background
RNAi experiments: Non-targeting RNAi (e.g., luciferase RNAi in C. elegans studies) provides the appropriate control for knockdown experiments
Rescue experiments: To confirm phenotype specificity:
Re-express RUBCN in knockout/knockdown models
Use RUBCN mutants lacking specific domains to map functional requirements
Apply tightly controlled expression systems to avoid overexpression artifacts
Tissue-specific considerations:
Temporal controls: For inducible systems:
Include vehicle-treated cohorts with the same genetic background
Perform time-course experiments to distinguish acute versus chronic effects
Consider age-dependent differences in RUBCN function
Methodological controls:
These control strategies ensure that observed phenotypes can be specifically attributed to RUBCN modulation rather than experimental artifacts or compensatory mechanisms.
Distinguishing between RUBCN expression levels and functional activity requires sophisticated experimental approaches:
Protein-protein interaction analysis:
Co-immunoprecipitation: Assess RUBCN binding to key partners like Beclin-1 or UVRAG
Proximity ligation assays: Visualize and quantify endogenous protein interactions in situ
FRET/BRET technology: Measure real-time interactions in living cells
Post-translational modification assessment:
Phosphorylation status: Use phospho-specific antibodies or mass spectrometry to detect activation-associated modifications
Ubiquitination analysis: Examine mono- versus poly-ubiquitination patterns that regulate RUBCN stability and function
Membrane association: Fractionate cells to distinguish cytosolic versus membrane-bound RUBCN pools
Functional readouts:
Autophagosome-lysosome fusion: Quantify colocalization of LC3 with LAMP1 to assess fusion events
Autophagic flux measurements: Use tandem fluorescent-tagged LC3 (mCherry-GFP-LC3) reporters to distinguish autophagosomes from autolysosomes
Lysosomal pH measurements: Assess whether RUBCN is affecting lysosomal acidification independent of fusion events
Real-time activity sensors:
Conformational biosensors: Design FRET-based tools to detect RUBCN conformational changes associated with activation
Optogenetic approaches: Use light-controllable RUBCN variants to directly test activity-function relationships
Domain-specific analysis:
Express RUBCN mutants lacking specific functional domains
Compare their effects on autophagy to distinguish which activities contribute to observed phenotypes
By combining these approaches, researchers can determine whether changes in autophagy during experimental conditions result from altered RUBCN expression levels or from post-translational modifications and protein interactions that modify its inhibitory activity on autophagosome-lysosome fusion.
RUBCN antibodies are increasingly deployed in metabolic disorder research with several innovative applications:
Tissue-specific metabolic profiling: Recent studies have revealed that RUBCN deficiency in kidney proximal tubular epithelial cells leads to metabolic syndrome with accelerated mobilization of phospholipids from cellular membranes to lysosomes . Conversely, hepatocyte-specific RUBCN deficiency improves liver steatosis and injury in high-fat diet models . RUBCN antibodies enable researchers to track these tissue-specific changes in protein expression and correlate them with metabolic outcomes.
Therapeutic target validation: As RUBCN inhibits autophagy, compounds designed to reduce its expression or activity represent potential therapeutic approaches for metabolic disorders characterized by impaired autophagy. HRP-conjugated RUBCN antibodies provide sensitive detection methods for high-throughput screening of such compounds.
Biomarker development: Changes in RUBCN levels may serve as biomarkers for metabolic disease progression or treatment response. Quantitative analysis using standardized RUBCN antibody-based assays helps establish these correlations.
Mechanistic insights: By combining RUBCN antibodies with metabolic flux analysis techniques, researchers have uncovered that RUBCN's effects on autophagy directly impact cellular metabolism through:
Altered lipid droplet turnover
Changed mitochondrial quality control
Disrupted membrane phospholipid recycling
Modified nutrient sensing pathways
Combinatorial therapeutic approaches: RUBCN antibodies help evaluate how autophagy modulation interacts with established metabolic disorder treatments, revealing synergistic or antagonistic effects that inform treatment strategies.
These applications demonstrate how RUBCN antibodies contribute to understanding the complex interplay between autophagy regulation and metabolic homeostasis, potentially leading to novel therapeutic interventions for conditions like obesity, diabetes, and non-alcoholic fatty liver disease.
Cutting-edge approaches for investigating RUBCN dynamics in living cells include:
Advanced fluorescent protein tagging:
Split fluorescent protein complementation: By tagging RUBCN and potential interaction partners with complementary fragments of fluorescent proteins, researchers can visualize protein interactions in real time
HaloTag and SNAP-tag systems: These self-labeling protein tags allow pulse-chase experiments to track RUBCN movement between subcellular compartments
Fluorescent timer proteins: These gradually change color over time, enabling determination of RUBCN protein age and turnover rates
Super-resolution microscopy techniques:
STED (Stimulated Emission Depletion): Achieves resolution below the diffraction limit, allowing visualization of RUBCN localization to specific membrane microdomains
PALM/STORM: Single-molecule localization methods that provide nanometer-scale resolution of RUBCN distribution
Lattice light-sheet microscopy: Enables long-term 3D imaging with minimal phototoxicity, ideal for tracking RUBCN dynamics during autophagy
Biosensor technologies:
FRET-based conformational sensors: Detect structural changes in RUBCN during activation/inactivation
FLIM (Fluorescence Lifetime Imaging): Measures subtle changes in protein environments independent of concentration
Optogenetic RUBCN variants: Light-activatable RUBCN constructs allow precise temporal control of its activity
Correlative light and electron microscopy (CLEM):
Combines fluorescence imaging of tagged RUBCN with electron microscopy's ultrastructural detail
Particularly valuable for studying RUBCN's association with autophagosomal and lysosomal membranes
Proximity labeling approaches:
BioID or APEX2 fusion proteins: When fused to RUBCN, these enzymes biotinylate proximal proteins, revealing the dynamic RUBCN interactome under different conditions
Split-BioID: Allows detection of specific protein-protein interactions in defined subcellular locations
These emerging methodologies provide unprecedented insights into RUBCN localization, interactions, and dynamics, enabling researchers to understand how this key autophagy regulator responds to various cellular stresses and metabolic conditions with high spatiotemporal resolution.
Translating RUBCN research from model organisms to human disease applications involves several critical considerations:
This translational framework ensures that fundamental discoveries about RUBCN biology in model organisms systematically advance toward clinically relevant applications, potentially leading to novel diagnostic and therapeutic approaches for autophagy-related human diseases.