RPS10P5 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship orders within 1-3 business days of receipt. Delivery times may vary depending on the order fulfillment method and destination. For specific delivery timeframes, please contact your local distributor.
Synonyms
RPS10P5 antibody; RPS10LPutative 40S ribosomal protein S10-like antibody
Target Names
RPS10P5
Uniprot No.

Q&A

What is RPS10P5 and why is it of interest to researchers?

The study of RPS10P5 using antibody-based approaches allows researchers to investigate potential non-canonical functions of this pseudogene, particularly if it produces RNA or protein products that might have biological significance. Understanding the expression patterns of RPS10P5 across different tissues and disease states could provide insights into its potential roles in normal physiology and pathological conditions.

How do antibodies against pseudogenes like RPS10P5 differ from typical protein-targeting antibodies?

Antibodies against pseudogene products present unique challenges compared to conventional protein-targeting antibodies. Since pseudogenes may have high sequence similarity to their parent genes, ensuring specificity is critical. When designing or selecting an RPS10P5 antibody, researchers must consider:

  • Epitope uniqueness: The antibody must target regions that differ from the related functional gene (RPS10)

  • Expression level considerations: Pseudogene products may be expressed at lower levels, requiring antibodies with higher sensitivity

  • Validation complexity: Additional controls are needed to confirm specificity against potential cross-reactivity with the parent gene products

  • Application optimization: Standard protocols may require modification for pseudogene detection

Unlike antibodies against abundantly expressed canonical proteins, pseudogene-targeting antibodies often require more rigorous validation through multiple complementary techniques to confirm their specificity and utility in research applications .

What basic validation should be performed before using RPS10P5 antibodies in research?

Before employing RPS10P5 antibodies in research projects, comprehensive validation is essential to ensure reliable results. A methodological approach should include:

  • Specificity testing:

    • Western blot analysis comparing RPS10P5 with RPS10 protein expression

    • Immunoprecipitation followed by mass spectrometry to confirm target identity

    • Testing in knockout/knockdown models where available

  • Application-specific validation:

    • For Western blotting: Confirm band size corresponds to predicted molecular weight

    • For immunohistochemistry: Include positive and negative control tissues

    • For flow cytometry: Compare with isotype controls and known expression patterns

  • Cross-reactivity assessment:

    • Testing on samples with different expression levels of RPS10 and related ribosomal proteins

    • Peptide competition assays to confirm epitope specificity

  • Lot-to-lot consistency verification:

    • Compare results between different antibody lots using standardized samples

    • Document batch information and validation results for reproducibility

Researchers should maintain detailed records of all validation experiments, as antibody performance can vary significantly between applications (WB, IHC, IF, Flow cytometry), which is particularly important when studying pseudogene products that may share sequence homology with functional proteins .

What is the optimal experimental design for studying RPS10P5 expression across different tissues?

When investigating RPS10P5 expression patterns across tissues, a robust experimental design should incorporate multiple complementary approaches:

Tissue Panel Analysis Protocol:

  • Sample selection and preparation:

    • Include diverse tissue types (minimum 6-8) with biological replicates (n≥3)

    • Prepare both fresh-frozen and FFPE samples for comprehensive analysis

    • Extract protein, RNA, and prepare tissue sections from the same specimens when possible

  • Multi-modal detection strategy:

    • Protein level: Western blot and immunohistochemistry with RPS10P5 antibody

    • RNA level: RT-qPCR with pseudogene-specific primers and RNA-seq analysis

    • In situ visualization: RNAscope or FISH for transcript localization

  • Essential controls:

    • Positive control: Tissues with known RPS10P5 expression

    • Negative control: Use of pre-immune serum or isotype controls

    • Specificity control: RPS10 expression analysis for comparison

  • Quantification approaches:

    • For IHC: Use standardized scoring system (H-score or digital image analysis)

    • For Western blot: Normalize to housekeeping proteins and include calibration curves

    • For RT-qPCR: Apply ΔΔCt method with carefully selected reference genes

This comprehensive approach helps overcome limitations of single-technique analyses and provides more reliable information about tissue-specific expression patterns of pseudogene products, which may be expressed at lower levels than conventional proteins .

How should researchers optimize immunohistochemistry protocols for RPS10P5 detection?

Optimizing immunohistochemistry (IHC) protocols for RPS10P5 detection requires systematic methodological refinement:

Optimized IHC Protocol Development:

  • Antigen retrieval optimization:

    • Test multiple methods (heat-induced vs. enzymatic)

    • Compare different buffers (citrate pH 6.0, EDTA pH 9.0, Tris-EDTA)

    • Adjust retrieval times (10-30 minutes) to maximize signal-to-noise ratio

    As demonstrated with other ribosomal proteins, heat-mediated antigen retrieval with Tris-EDTA buffer (pH 9.0) for 10 minutes often yields optimal results .

  • Antibody concentration titration:

    • Perform serial dilutions (recommended starting range: 1:50 to 1:1000)

    • Assess both signal intensity and background at each concentration

    • Select optimal dilution with maximum specific signal and minimal background

  • Detection system selection:

    • Compare polymer-based systems with conventional avidin-biotin methods

    • Test amplification systems for low-abundance targets

    • Evaluate chromogen options (DAB vs. alternatives) for optimal visualization

  • Incubation parameters:

    • Test both short (1-2h) and overnight primary antibody incubations at 4°C

    • Optimize blocking conditions to reduce non-specific binding

    • Adjust washing steps to minimize background

  • Validation through controls:

    • Peptide competition assays to confirm specificity

    • Comparison with RNA expression data (ISH or RNA-seq)

    • Testing on tissues with known expression patterns

Standardizing and documenting these optimization steps ensures reproducible detection of potentially low-abundance pseudogene products across different experimental conditions .

What approaches can effectively distinguish between RPS10P5 and RPS10 in experimental settings?

Distinguishing between RPS10P5 and its related functional gene RPS10 presents a significant challenge due to potential sequence similarities. Researchers should employ a multi-faceted approach:

Differential Detection Strategy:

  • Targeted antibody selection:

    • Use antibodies specifically raised against unique epitopes in RPS10P5

    • Perform epitope mapping to confirm antibody binding sites

    • Consider generating custom antibodies against divergent regions

  • Nucleic acid-based discrimination:

    • Design primers/probes targeting pseudogene-specific variations

    • Utilize droplet digital PCR for absolute quantification

    • Employ RNA-seq with computational pipelines specific for pseudogene detection

  • Functional differentiation:

    • Knockout/knockdown studies targeting each gene individually

    • Complementation experiments to assess functional differences

    • Protein-protein interaction studies to identify differential binding partners

  • Subcellular localization analysis:

    • Compare localization patterns through immunofluorescence

    • Perform subcellular fractionation followed by Western blotting

    • Use proximity ligation assays to detect potential co-localization or separation

This comprehensive approach integrates molecular, biochemical, and cellular techniques to provide complementary evidence for distinguishing between the pseudogene and its functional counterpart, which is essential for accurate interpretation of experimental results .

How does fixation affect RPS10P5 antibody staining and what protocols can mitigate these effects?

Fixation can significantly impact antibody staining for targets like RPS10P5, with effects varying based on the specific epitope and antibody characteristics. Research indicates that fixation can lead to either signal enhancement or reduction depending on the target:

Fixation Effects and Mitigation Strategies:

Fixation MethodTypical Effect on EpitopesRecommended Mitigation
Formalin (4%, 24h)May mask epitopes through protein cross-linkingExtended antigen retrieval (15-20 min) with Tris-EDTA (pH 9.0)
Paraformaldehyde (2%, 1h)Moderate epitope maskingUse retrieval buffer specific to the epitope location (membrane vs. cytoplasmic)
Methanol (-20°C, 10 min)Preserves some epitopes but can denature othersTest with and without additional retrieval steps
Acetone (4°C, 10 min)Minimal epitope masking but poor morphologyConsider for flow cytometry applications
Fresh-frozenMinimal epitope alterationRecommended for initial antibody characterization

As observed in comprehensive antibody staining studies, certain markers show significant fixation-dependent signal variation . For RPS10P5 detection, researchers should:

  • Compare multiple fixation methods during antibody validation

  • Document fixation-dependent changes in staining patterns

  • Standardize fixation protocols once optimal conditions are identified

  • Consider dual approaches (fixed and unfixed) for comprehensive analysis

Importantly, testing a panel of antigen retrieval conditions is essential, as some epitopes demonstrate recovery-dependent variations that significantly impact detection sensitivity and specificity .

What are the best practices for multiplexing RPS10P5 antibody with other markers?

Multiplexed detection of RPS10P5 alongside other markers provides valuable contextual information about its expression. Implementing best practices for multiplexing ensures reliable results:

Multiplexing Optimization Protocol:

  • Panel design considerations:

    • Select markers with non-overlapping subcellular localization when possible

    • Choose primary antibodies from different host species to avoid cross-reactivity

    • Include markers for cellular context (lineage, activation state, etc.)

  • Technical optimization:

    • Perform sequential staining if antibodies require different retrieval conditions

    • Titrate each antibody in the multiplex panel independently, then together

    • Include appropriate blocking steps between antibody applications

  • Signal separation strategies:

    • For fluorescence: Select fluorophores with minimal spectral overlap

    • For chromogenic detection: Use distinct chromogens with good visual separation

    • Apply spectral unmixing algorithms for closely related signals

  • Controls for multiplexed detection:

    • Single-stained controls for each antibody

    • Fluorescence minus one (FMO) controls for flow cytometry

    • Absorption controls for chromogenic IHC

Mass cytometry approaches can be particularly valuable for complex multiplexing, allowing simultaneous detection of up to 40 parameters with minimal signal overlap, which is especially useful when studying pseudogene products in the context of broader cellular phenotypes .

What quantitative analysis approaches are most appropriate for RPS10P5 antibody-based studies?

Quantitative analysis of RPS10P5 antibody staining requires rigorous analytical approaches tailored to the detection method:

Quantitative Analysis Framework:

  • Western blot quantification:

    • Use calibration curves with recombinant standards when available

    • Apply digital image analysis with background subtraction

    • Normalize to multiple housekeeping proteins for reliable quantification

    • Report results as fold-change relative to controls with statistical analysis

  • Immunohistochemistry quantification:

    • Apply digital pathology approaches with automated tissue segmentation

    • Use H-score (intensity × percentage) or Allred scoring systems

    • Implement machine learning algorithms for pattern recognition

    • Report both intensity and distribution parameters

  • Flow cytometry analysis:

    • Establish positive populations using FMO controls

    • Report both percentage positive and median fluorescence intensity

    • Apply dimensionality reduction techniques (tSNE, UMAP) for complex datasets

    • Use standardized beads for inter-experimental calibration

  • Mass cytometry data analysis:

    • Implement unsupervised clustering algorithms

    • Apply Earth Mover's Distance or other metrics for population comparison

    • Use trajectory analysis for developmental or activation studies

    • Integrate with transcriptomic data when available

These quantitative approaches should incorporate appropriate statistical methods, including normality testing, selection of parametric or non-parametric tests, and correction for multiple comparisons. For longitudinal studies, mixed-effects models may be more appropriate than repeated measures ANOVA .

How should researchers address non-specific binding or high background when using RPS10P5 antibodies?

Non-specific binding and high background are common challenges with antibodies targeting low-abundance targets like pseudogene products. A systematic troubleshooting approach includes:

Background Reduction Protocol:

  • Antibody-specific optimizations:

    • Further dilute primary antibody (test 2-5× higher dilutions)

    • Reduce incubation time or temperature

    • Try different antibody clones if available

    • Use affinity-purified antibodies when possible

  • Blocking optimizations:

    • Test alternative blocking reagents (BSA, normal serum, commercial blockers)

    • Extend blocking time (1-2 hours or overnight)

    • Include protein additives in antibody diluent (0.1-0.5% BSA)

    • Consider adding 0.1-0.3% Triton X-100 for intracellular targets

  • Washing protocol refinements:

    • Increase number and duration of washing steps

    • Use detergent-containing wash buffers (0.05-0.1% Tween-20)

    • Apply gentle agitation during washing

    • Consider higher salt concentration in wash buffer (150-500 mM NaCl)

  • Signal-to-noise enhancement:

    • Use biotinylated secondary antibodies with streptavidin-HRP for signal amplification

    • Consider tyramide signal amplification for very low abundance targets

    • Apply Sudan Black B (0.1-0.3%) to reduce autofluorescence in fluorescent applications

    • Use antigen retrieval optimization to enhance specific signal

For histological applications specifically, tissue preparation factors such as proper fixation timing and thorough deparaffinization can significantly reduce background staining .

What controls are essential for validating RPS10P5 antibody specificity in different applications?

Rigorous validation of RPS10P5 antibody specificity requires comprehensive controls tailored to each application:

Application-Specific Control Panel:

ApplicationEssential ControlsPurpose
Western Blot- Recombinant protein standard
- Knockdown/knockout lysates
- Peptide competition
- Related protein samples (RPS10)
Confirms molecular weight and specificity; identifies potential cross-reactive species
IHC/ICC- Known positive tissue
- Negative tissue
- Isotype control
- Peptide blocking
- Secondary-only control
Verifies staining pattern and evaluates non-specific binding sources
Flow Cytometry- FMO controls
- Isotype controls
- Unstained cells
- Cells with/without target expression
- Titration series
Establishes gating boundaries and determines optimal signal separation
IP/Co-IP- Input control
- IgG control
- Reverse IP
- Stringency controls
Confirms specificity of pulled-down proteins and evaluates interaction strength

For pseudogene products like RPS10P5, additional controls should include:

  • Parallel detection of the related functional gene (RPS10)

  • RNA-level validation (RT-qPCR or RNA-seq) to correlate with protein detection

  • Spatial correlation controls (IF/IHC with RNA-FISH) to verify co-localization of protein with transcript

These comprehensive controls help distinguish true target detection from artifacts, which is particularly important for potentially low-abundance pseudogene products .

What are the most common causes of experimental variability in RPS10P5 antibody-based assays and how can they be minimized?

Experimental variability in antibody-based assays for targets like RPS10P5 can arise from multiple sources. Identifying and mitigating these factors is essential for reproducible research:

Sources of Variability and Mitigation Strategies:

  • Antibody-related variability:

    • Issue: Lot-to-lot inconsistency in commercial antibodies

    • Mitigation: Purchase sufficient quantity from single lot for entire study; validate each new lot against previous standards; document lot numbers in publications

  • Sample preparation variability:

    • Issue: Inconsistent fixation, extraction, or processing conditions

    • Mitigation: Standardize all preparation protocols with precise timing, temperature, and reagent specifications; process all experimental groups in parallel

  • Technical execution variability:

    • Issue: Differences in handling between operators or experiments

    • Mitigation: Develop detailed SOPs; use automated systems where possible; implement technical training and competency assessment; include technical replicates

  • Instrument-related variability:

    • Issue: Fluctuations in instrument performance over time

    • Mitigation: Regular calibration; include standard samples in each experiment; use internal reference standards; document instrument settings

  • Analysis-related variability:

    • Issue: Subjective interpretation or inconsistent quantification approaches

    • Mitigation: Use automated analysis pipelines; blind the analyst to experimental groups; establish quantification criteria before beginning analysis

The implementation of quality control metrics throughout the experimental workflow significantly reduces variability. For longitudinal studies, preparing a large batch of control samples for use across multiple experiments provides an essential reference point for normalizing data .

How can RPS10P5 antibodies be utilized in single-cell analysis techniques?

Single-cell analysis with RPS10P5 antibodies offers unique insights into heterogeneous expression patterns. Implementation requires specialized approaches:

Single-Cell Analysis Implementation:

  • Mass cytometry (CyTOF) applications:

    • Metal-conjugate the RPS10P5 antibody using commercial kits

    • Include RPS10P5 in panels with lineage markers and functional indicators

    • Apply unsupervised clustering algorithms to identify correlated expression patterns

    • Integrate with single-cell transcriptomics when possible

  • Single-cell Western blotting:

    • Optimize lysis conditions for individual cells

    • Establish detection limits for low-abundance targets

    • Use enhanced chemiluminescence or fluorescent detection for sensitivity

    • Compare RPS10P5 with RPS10 expression at single-cell level

  • Imaging mass cytometry or multiplexed immunofluorescence:

    • Design panels that include RPS10P5 alongside tissue architecture markers

    • Apply computational spatial analysis for neighborhood relationships

    • Quantify subcellular localization patterns

    • Correlate with functional or disease phenotypes

  • Suspension mass cytometry workflow optimization:

    • Test fixation conditions that preserve epitopes

    • Optimize permeabilization for intracellular detection

    • Use barcoding strategies for batch consistency

    • Apply two-tiered barcoding for large-scale experiments

These single-cell approaches provide critical information about cellular heterogeneity and can reveal subpopulations with distinct RPS10P5 expression patterns that might be masked in bulk analyses .

What are the considerations for using RPS10P5 antibodies in bispecific antibody development?

Developing bispecific antibodies incorporating anti-RPS10P5 binding domains presents unique challenges and opportunities:

Bispecific Development Considerations:

  • Format selection based on application:

    • For imaging: Smaller formats (diabodies, single-chain bispecifics)

    • For functional modulation: IgG-like formats with Fc functionality

    • For targeting: Anti-RPS10P5 × anti-effector cell receptor formats

  • Molecular design optimization:

    • Engineer stable heterodimerization through knob-into-hole or other technologies

    • Select optimal linker length and composition for proper domain orientation

    • Incorporate stabilizing mutations if needed for thermal stability

    • Consider the spatial arrangement of binding domains based on target biology

  • Production and purification challenges:

    • Establish screening systems for correct heavy chain pairing

    • Implement purification strategies to remove incorrectly paired species

    • Validate binding to both targets independently and simultaneously

    • Assess stability under physiological conditions

  • Functional validation:

    • Test binding kinetics to both targets using surface plasmon resonance

    • Verify simultaneous binding capability through bridging assays

    • Evaluate functional activity in relevant biological systems

    • Assess potential immunogenicity of novel junctions

The increased structural complexity of bispecific antibodies incorporating anti-RPS10P5 domains requires judicious design considerations and extensive molecular engineering to ensure formation of high-quality bispecifics with the intended mode of action and favorable drug-like qualities .

How can researchers integrate RPS10P5 antibody data with other -omics approaches for comprehensive analysis?

Integrating RPS10P5 antibody-based data with complementary -omics approaches provides a more comprehensive understanding of biological systems:

Multi-omics Integration Strategy:

  • Experimental design for integrated analysis:

    • Collect matched samples for parallel -omics analyses

    • Include appropriate time points to capture dynamic processes

    • Design consistent perturbation strategies across platforms

    • Incorporate sufficient biological replicates (n≥3 for each condition)

  • Complementary data generation:

    • Transcriptomics: RNA-seq with specific analysis of pseudogene transcripts

    • Proteomics: Mass spectrometry with targeted approaches for low-abundance species

    • Epigenomics: ChIP-seq or ATAC-seq to assess regulatory mechanisms

    • Interactomics: IP-MS to identify RPS10P5 binding partners if expressed

  • Data integration approaches:

    • Correlation analysis across platforms (protein vs. transcript)

    • Network analysis to identify functional modules

    • Causal modeling to establish regulatory relationships

    • Machine learning approaches for pattern recognition

  • Visualization and interpretation:

    • Generate integrated heatmaps with hierarchical clustering

    • Develop multi-dimensional visualizations (e.g., using dimension reduction)

    • Apply pathway enrichment to identify biological processes

    • Create interactive dashboards for exploratory analysis

This integrated approach can reveal relationships between RPS10P5 expression and broader cellular processes, potentially uncovering functional roles for this pseudogene that would not be apparent from antibody-based studies alone .

What are the emerging technologies that might enhance RPS10P5 detection and functional characterization?

Several cutting-edge technologies hold promise for advancing RPS10P5 research:

  • Proximity labeling approaches:

    • BioID or APEX2 fusion proteins to identify spatial interaction partners

    • Proximity extension assays for ultrasensitive detection

    • CITE-seq for combined protein and transcript detection at single-cell level

  • Advanced imaging techniques:

    • Super-resolution microscopy for subcellular localization

    • Live-cell imaging with tagged antibody fragments

    • Expansion microscopy for improved spatial resolution

    • 4D imaging for temporal dynamics of expression

  • Functional genomics approaches:

    • CRISPR activation/interference for pseudogene modulation

    • RNA-targeting CRISPR systems for transcript-specific perturbation

    • Massively parallel reporter assays for regulatory element identification

    • Synthetic biology approaches to assess potential functions

  • Computational advances:

    • Deep learning for image analysis and pattern recognition

    • Advanced algorithms for pseudogene-specific transcript quantification

    • Systems biology modeling incorporating pseudogene regulation

    • Knowledge graph approaches for literature mining and hypothesis generation

These emerging technologies will enable more sensitive detection and more comprehensive functional characterization of pseudogenes like RPS10P5, potentially revealing unexpected biological roles and disease associations .

What gaps remain in our understanding of RPS10P5 and how might new antibody technologies address them?

Despite advances in antibody technologies, several knowledge gaps remain in RPS10P5 research:

  • Expression pattern characterization:

    • Comprehensive tissue and cell-type specific expression maps are lacking

    • Developmental regulation remains largely unexplored

    • Disease-associated expression changes need systematic investigation

    • Subcellular localization patterns require detailed characterization

  • Functional significance:

    • Potential non-canonical functions are poorly understood

    • Regulatory relationships with RPS10 and other genes remain speculative

    • Contribution to normal physiology or disease pathogenesis is unclear

    • Evolutionary conservation and species-specific differences need exploration

  • Technological challenges:

    • Detection sensitivity limits for potentially low-abundance products

    • Specificity issues due to sequence similarity with functional genes

    • Limited availability of validated research tools

    • Standardization of detection and quantification approaches

New antibody technologies that could address these gaps include:

  • Recombinant antibodies with enhanced specificity and batch consistency

  • Nanobodies or single-domain antibodies for improved tissue penetration

  • Antibody-oligonucleotide conjugates for ultrasensitive detection

  • Cyclic immunofluorescence for highly multiplexed tissue analysis

Addressing these gaps will require collaborative efforts across multiple research groups, using complementary approaches and standardized reporting to build a comprehensive understanding of RPS10P5 biology .

How can standardization in RPS10P5 antibody research improve reproducibility and accelerate discoveries?

Standardization in antibody-based research for targets like RPS10P5 is critical for improving reproducibility and accelerating scientific progress:

Standardization Framework:

  • Reagent standardization:

    • Implement consistent antibody validation criteria across labs

    • Establish biobanks of reference materials and control samples

    • Create openly accessible standard operating protocols

    • Develop community-wide antibody validation repositories

  • Methodology standardization:

    • Adopt common experimental workflows with defined parameters

    • Implement consistent reporting formats for methods sections

    • Establish minimum information guidelines for antibody experiments

    • Create analytical pipelines that can be shared between researchers

  • Data standardization:

    • Use consistent data formats and metadata annotations

    • Deposit raw data in public repositories with detailed protocols

    • Implement FAIR (Findable, Accessible, Interoperable, Reusable) principles

    • Develop standardized quality metrics for data evaluation

  • Reporting standardization:

    • Adopt comprehensive checklist-based reporting (e.g., ARRIVE guidelines)

    • Implement structured methods sections with standardized terminology

    • Include detailed reagent information including catalog numbers and lot numbers

    • Report both positive and negative results to address publication bias

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.