OSBPL2 binds sterols and phospholipids, associating with the Golgi apparatus and lipid droplets . Its disruption has been linked to:
Hearing loss: OSBPL2 mutations cause autosomal dominant non-syndromic hearing loss (ADNSHL), with studies showing its role in maintaining auditory cell survival via AKT/FOXG1 signaling .
Lipid metabolism: Interactions with ATIC modulate AMPK signaling, impacting cholesterol biosynthesis and energy metabolism .
Obesity: Deletion accelerates preadipocyte differentiation, suggesting a role in adipogenesis .
Antibodies targeting OSBPL2 would likely be used in:
Immunohistochemistry: To localize OSBPL2 in tissues like the inner ear or adipose tissue .
Western blotting: For protein expression analysis in studies of hearing loss or metabolic disorders .
ELISA assays: To quantify OSBPL2 levels in cellular lysates or bodily fluids .
The absence of direct antibody data in the provided sources highlights a gap in antibody-specific studies. Key challenges include:
Antibody specificity: Ensuring cross-reactivity with truncated/mutated OSBPL2 variants, as mutations (e.g., p.His60Glnfs*93) lead to protein aggregation .
Tissue distribution: Optimizing antibodies for detection in lipid-rich environments or auditory cells .
OSBPL2 (oxysterol binding protein-like 2, also known as ORP2) is a member of the oxysterol-binding protein family involved in lipid transport and signaling. Current research focuses on:
Hearing loss mechanisms, particularly DFNA67 autosomal dominant hearing loss
Inner and outer hair cell stereocilia development
Ciliogenesis and Sonic hedgehog (Shh) signaling
Lipid metabolism and transport
OSBPL2 has a calculated molecular weight of 55 kDa, though observed weights of 51 kDa and 56 kDa have been reported in Western blot applications . The protein contains two main domains: the FFAT (two phenylalanines in an acidic tract) motif, which targets the endoplasmic reticulum, and the ORD (OSBP-related domain), essential for binding and transferring sterols, oxysterols, and phosphoinositides .
For optimal Western blot detection of OSBPL2:
Sample preparation:
Gel electrophoresis:
Load 20-30 μg of total protein per lane
Run on 10-12% SDS-PAGE gel
Transfer and blocking:
Transfer to PVDF membrane
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Antibody incubation:
Detection:
Controls:
Recommended dilutions vary by application and specific antibody. For antibody 14751-1-AP:
| Application | Recommended Dilution | Notes |
|---|---|---|
| Western Blot (WB) | 1:1000-1:8000 | Optimize based on your specific sample |
| Immunoprecipitation (IP) | 0.5-4.0 μg per 1.0-3.0 mg lysate | Effective for HeLa cell lysates |
| Immunohistochemistry (IHC) | 1:50-1:500 | Use TE buffer pH 9.0 for antigen retrieval |
| Immunofluorescence (IF-P) | 1:200-1:800 | Validated in mouse brain tissue |
For other antibodies:
DyLight 650-conjugated antibody (NBP2-97886C): Optimal dilution should be experimentally determined for IHC-P
DF12681: Recommended for WB and IF/ICC applications in human and mouse samples
C-terminal antibody (ABIN185116): Effective in ELISA (detection limit dilution 1:8000)
Always perform a dilution series to optimize for your specific experimental conditions and sample types.
Validating antibody specificity using knockout models is crucial for ensuring reliable results:
Generate OSBPL2 knockout models:
Validation strategy:
Western blot: Compare wild-type vs. knockout samples
Look for absence of bands at 51-56 kDa in knockout samples
Include loading controls (β-actin, GAPDH)
Test across multiple cell lines when possible (HEK293, HEI-OC1, HeLa)
Additional controls:
Quantitative validation:
For researchers studying OSBPL2 in hearing loss models, consider validating with both homozygous and heterozygous knockout mice, as phenotypes may differ between genotypes .
Detecting wild-type and mutant OSBPL2 proteins in the same sample presents unique challenges:
Understanding mutant properties:
Subcellular localization analysis:
Use immunofluorescence to compare distribution patterns
Wild-type OSBPL2: Diffusely distributed throughout cytoplasm
Truncated mutants: Form distinct cytoplasmic aggregates
Protocol:
Fix cells in 4% paraformaldehyde (10 min, RT)
Permeabilize with 0.1% Triton X-100 (3 min, RT)
Block with 1% BSA in PBS
Incubate with primary antibody overnight at 4°C
Detect with appropriate fluorophore-conjugated secondary antibodies
Western blot considerations:
Protein stability assessment:
Co-immunoprecipitation:
Cochlear tissue presents unique challenges for immunofluorescence due to its complex structure:
Tissue preparation:
Staining protocol optimization:
Thaw and permeabilize sections with 0.1% Triton X-100 (3 min, RT)
Block with 1% BSA in PBS
For OSBPL2 detection, recommended antibody dilution: 1:50 (goat anti-ORP-2)
For co-localization studies, combine with markers such as:
Anti-prestin (1:5000) for outer hair cells
Anti-otoferlin (1:10000) for inner hair cells
Incubate overnight at 4°C
Detect with appropriate secondary antibodies (Cy3 or Alexa488-conjugated)
Mount with Vectashield containing DAPI
Imaging considerations:
Controls and validation:
Include OSBPL2 knockout tissue as negative control
Use double-staining with known hair cell markers to confirm localization
Compare wild-type and mutant expression patterns in cochlear sections
Co-immunoprecipitation (Co-IP) is valuable for studying OSBPL2 protein interactions:
Experimental design:
For exogenous expression: Use tagged constructs (FLAG-OSBPL2) in HEK293T cells
For endogenous interactions: Use specific OSBPL2 antibodies with appropriate cell/tissue lysates
Protocol optimization:
Lysis buffer: 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, with protease inhibitors
For membrane-associated interactions, consider adding 0.5% sodium deoxycholate
Clear lysates by centrifugation (14,000 × g, 10 min, 4°C)
Pre-clear with protein A/G beads to reduce non-specific binding
Antibody selection:
Washing and elution:
Wash beads at least 5 times with lysis buffer
Elute in 6× SDS loading buffer by boiling (100°C, 10 min)
Analyze by Western blotting with appropriate antibodies
Controls and validation:
Advanced applications:
Inconsistent OSBPL2 detection across different biological samples can be addressed systematically:
Expression level variations:
Antibody selection considerations:
Sample preparation optimization:
For membrane-associated OSBPL2, include mild detergents (0.5% NP-40)
For aggregation-prone mutants, adjust lysis conditions appropriately
Sample storage: Avoid multiple freeze-thaw cycles
Protocol adjustments:
Systematic optimization approach:
Test multiple antibody dilutions (1:500, 1:1000, 1:2000, 1:4000, 1:8000)
Compare different detection systems (ECL vs. fluorescent)
Optimize incubation times and temperatures
Validation strategies:
Confirm specificity using siRNA knockdown or CRISPR/Cas9 knockout
Verify transcript levels by RT-PCR or RT-qPCR
Consider exogenous expression of tagged OSBPL2 as positive control
Research on OSBPL2 mutations in hearing loss requires rigorous controls:
Genetic model controls:
Phenotypic assessments:
Perform auditory brainstem response (ABR) testing at multiple frequencies
Compare ABR thresholds across genotypes and age groups
Assess distortion product otoacoustic emissions (DPOAEs)
Molecular analysis controls:
Histological examinations:
Compare cochlear morphology between genotypes
Use double immunofluorescence with markers for:
Hair cells (prestin, otoferlin)
Stereocilia (phalloidin)
Primary cilia markers if studying ciliogenesis
Functional studies:
For lipid metabolism studies, include serum lipid profile analysis
For ciliary function, analyze Shh pathway activation using GLI3 localization
For autophagy studies, monitor LC3 conversion with/without inhibitors
Rescue experiments:
Recent research has established OSBPL2's role in ciliogenesis:
Experimental approaches:
Generate OSBPL2-deficient cells using CRISPR/Cas9
Compare ciliary length and structure in wild-type vs. knockout cells
Analyze phosphoinositide distribution on ciliary membranes
Key experimental findings:
Phosphoinositide analysis:
PI(4,5)P₂ levels on ciliary membranes are regulated by OSBPL2
Methods to study PI distribution include specialized immunofluorescence techniques
Sonic hedgehog (Shh) signaling assessment:
Recommended protocols:
Serum-starve cells (24h) to induce cilia formation
Immunostain for acetylated α-tubulin to visualize cilia
Measure cilia length and quantify percentage of ciliated cells
Analyze ciliary localization of Shh pathway components after SAG treatment
OSBPL2 mutations can lead to protein aggregation and autophagy defects:
Protein aggregation analysis:
Compare wild-type and mutant OSBPL2 localization by immunofluorescence
Observe formation of cytoplasmic aggregates with truncated mutants
Use Western blot under denaturing conditions to detect multimers
Lysosomal degradation assessment:
Protein stability experiments:
Perform cycloheximide chase assays (100 μg/ml)
Collect samples at multiple timepoints (0, 3, 6, 9, 12h)
Compare degradation rates between wild-type and mutant proteins
Autophagy flux assessment:
Monitor LC3-I to LC3-II conversion by Western blot
Use tandem mRFP-GFP-LC3 constructs to differentiate autophagosome formation vs. maturation
Compare basal vs. stress-induced autophagy (starvation, rapamycin treatment)
Protein-protein interaction analysis: