RPL37A Antibody

Shipped with Ice Packs
In Stock

Description

Overview of RPL37A Antibody

RPL37A antibodies are polyclonal reagents primarily raised in rabbits, targeting the RPL37A protein (UniProt ID: P61513). These antibodies are validated for use in Western blot (WB), immunohistochemistry (IHC), immunoprecipitation (IP), immunofluorescence (IF), and flow cytometry (FC) .

PropertyDetails
TargetRPL37A (ribosomal protein L37a)
Host SpeciesRabbit
ReactivityHuman, Mouse, Rat
Molecular WeightObserved: 10 kDa (Proteintech) ; Calculated: 10.3 kDa (Boster Bio)
ImmunogenFusion protein (Proteintech) or synthetic peptide (Boster Bio)
Key ApplicationsWB, IHC, IF, IP, FC

Research Applications and Validation Data

RPL37A antibodies are utilized across diverse experimental contexts:

2.1. Key Applications

ApplicationConditions
Western BlotDilution: 1:500–1:4000; Detected in HeLa, MCF-7, SH-SY5Y cells
ImmunohistochemistryDilution: 1:20–1:200; Validated in human breast cancer tissue
ImmunofluorescenceDilution: 1:20–1:200; Confirmed in SH-SY5Y cells
Flow Cytometry0.20 µg/10^6 cells; Tested in MCF-7 cells

2.2. Clinical Relevance

A 2021 PubMed study highlighted RPL37A's role as a prognostic biomarker in locally advanced breast cancer (LABC) :

  • High RPL37A expression correlated with improved pathological complete response (PCR) to neoadjuvant chemotherapy (NCT) (OR = 2.1, p < 0.05) .

  • Low RPL37A levels were associated with increased recurrence risk (HR = 1.8) and cancer-specific mortality (HR = 2.3) .

Technical Considerations

  • Storage: Stable at -20°C for 1 year; avoid freeze-thaw cycles .

  • Antigen Retrieval: For IHC, use TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

  • Controls: Jurkat, MCF-7, and HeLa lysates serve as positive controls .

Research Implications

RPL37A antibodies have advanced understanding of ribosomal biology and disease mechanisms:

  • Cancer Biomarker: High RPL37A expression predicts favorable NCT response in breast cancer .

  • Therapeutic Target: Dysregulated RPL37A is linked to oncogenesis, suggesting potential for ribosomal-targeted therapies .

Limitations and Future Directions

  • Species Cross-Reactivity: Limited data beyond human, mouse, and rat .

  • Functional Studies: Further research is needed to clarify RPL37A's role in metastasis and drug resistance .

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on your location and the shipping method chosen. For specific delivery timeframes, please contact your local distributor.
Synonyms
RPL37A antibody; 60S ribosomal protein L37a antibody; Large ribosomal subunit protein eL43 antibody
Target Names
RPL37A
Uniprot No.

Q&A

What is RPL37A and why is it significant in cellular research?

RPL37A (ribosomal protein L37a) is a component of the 60S ribosomal subunit with a calculated molecular weight of 10 kDa . Beyond its canonical role in protein synthesis, RPL37A has emerged as a significant regulatory protein in the Mdm2-p53 pathway, contributing to cell cycle regulation and apoptotic responses . Recent studies have identified RPL37A as a potential biomarker for response to neoadjuvant chemotherapy (NCT) in breast cancer patients, highlighting its clinical relevance . Investigating RPL37A function through antibody-based techniques provides insights into both fundamental ribosomal biology and disease mechanisms, particularly in cancer research.

Which experimental applications are validated for commercially available RPL37A antibodies?

Currently available RPL37A antibodies have been validated for multiple experimental applications, with consistent performance across several techniques:

ApplicationValidated Dilution RangesPositive Detection Examples
Western Blot (WB)1:500-1:5000MCF-7, HeLa, SH-SY5Y, COLO 320 cells
Immunohistochemistry (IHC)1:20-1:300Human breast cancer tissue
Immunofluorescence (IF/ICC)1:20-1:200SH-SY5Y cells
Flow Cytometry (FC, Intracellular)0.20 μg per 10^6 cellsMCF-7 cells
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg lysateMCF-7 cells
ELISAUp to 1:40000Various samples

The antibody has demonstrated consistent reactivity with human, mouse, and rat samples , though cross-reactivity with other species may require additional validation.

How should RPL37A antibodies be stored and handled to maintain optimal performance?

For optimal performance of RPL37A antibodies:

  • Long-term storage: Maintain at -20°C where the antibody remains stable for approximately one year after shipment .

  • Short-term/frequent use: Store at 4°C for up to one month to minimize freeze-thaw cycles .

  • Avoid repeated freeze-thaw cycles as these can compromise antibody integrity and performance .

  • Most commercial preparations contain 50% glycerol with stabilizers (0.02% sodium azide, sometimes with 0.1% BSA in smaller aliquots) , eliminating the need for additional aliquoting prior to -20°C storage.

  • When working with the antibody, allow it to equilibrate to room temperature before opening to prevent condensation that could introduce contaminants.

Proper storage management significantly impacts experimental reproducibility and maintains the functional lifetime of the antibody.

How should RPL37A antibody dilutions be optimized for Western blot applications in different cell types?

Optimization of RPL37A antibody for Western blot requires systematic titration across different cell types:

  • Initial range finding: Begin with the manufacturer's recommended range (typically 1:1000-1:4000) using positive control lysates (e.g., MCF-7 or SH-SY5Y cells) where RPL37A expression is established.

  • Systematic titration: Prepare a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:4000) to identify optimal signal-to-noise ratio. Load equivalent amounts of total protein (20-30 μg) per lane.

  • Cell-type optimization: Different cell lines may require adjusted dilutions:

    • Cancer cell lines (MCF-7, HeLa): Often require less concentrated antibody (1:2000-1:4000)

    • Primary cells: May require more concentrated antibody (1:500-1:1000)

  • Detection system considerations: Adjust dilutions based on whether using chemiluminescence (more sensitive) or colorimetric detection (less sensitive).

  • Validation criteria: The optimal dilution should produce:

    • Clear band at 10 kDa (calculated molecular weight of RPL37A)

    • Minimal background/non-specific binding

    • Linear response relative to protein loading

For challenging samples, incorporating a blocking peptide control can verify specificity of detected bands .

What are the critical parameters for successful RPL37A detection in paraffin-embedded tissue samples?

Successful RPL37A detection in paraffin-embedded tissues requires attention to several critical parameters:

  • Antigen retrieval: RPL37A antibodies typically require heat-induced epitope retrieval:

    • Primary recommendation: TE buffer pH 9.0

    • Alternative method: Citrate buffer pH 6.0 or Tris-EDTA with high-pressure/temperature treatment

  • Antibody concentration: For paraffin sections, use more concentrated dilutions (1:20-1:200) than for Western blot.

  • Incubation conditions: Optimal detection typically requires overnight incubation at 4°C .

  • Blocking: Pre-incubate sections with appropriate blocking buffer containing serum matched to the secondary antibody host.

  • Validation controls:

    • Negative control: Primary antibody pre-absorbed with immunogen peptide

    • Positive control: Human breast cancer tissue has demonstrated consistent RPL37A expression

  • Signal amplification: For low abundance detection, consider using polymer-based detection systems over traditional ABC methods.

  • Counterstain selection: Hematoxylin works well but avoid overstaining which can mask specific signals.

The data indicates that RPL37A antibodies perform most consistently in breast cancer tissues, making these excellent positive controls for method optimization .

What experimental conditions influence RPL37A subcellular localization detection in immunofluorescence applications?

RPL37A subcellular localization can be influenced by multiple experimental factors in immunofluorescence applications:

  • Fixation method:

    • For consistent RPL37A detection, -20°C ethanol fixation has shown good results with SH-SY5Y cells

    • Paraformaldehyde (4%) may preserve structure but might mask some RPL37A epitopes

  • Cell permeabilization:

    • Required for intracellular detection

    • Gentle permeabilization (0.1-0.2% Triton X-100) preferred to preserve ribosomal structures

  • Antibody dilution:

    • Start with 1:50-1:100 dilution for IF applications

    • May require more concentrated antibody than Western blot

  • Secondary antibody selection:

    • Anti-rabbit IgG conjugated to bright fluorophores (e.g., Alexa Fluor 488) provides optimal visualization

    • Control for cross-reactivity with other cellular components

  • Confocal microscopy settings:

    • RPL37A typically shows predominantly cytoplasmic localization with possible nucleolar enrichment

    • Z-stack imaging may be necessary to fully capture distribution

  • Co-localization studies:

    • Consider dual staining with nucleolar markers (fibrillarin, nucleolin) or ribosomal markers

    • Appropriate channel separation required to avoid bleed-through

When studying RPL37A translocation during stress responses, careful time-course experiments with consistent imaging parameters are essential to detect subtle changes in localization patterns.

How can researchers differentiate between true RPL37A signal and non-specific binding in Western blot applications?

Differentiating true RPL37A signal from non-specific binding requires multiple validation approaches:

  • Molecular weight verification:

    • RPL37A should appear at approximately 10 kDa

    • Some reports show observation at up to 72 kDa , which may represent post-translational modifications or oligomeric forms

  • Positive controls:

    • Include lysates from cells known to express RPL37A (MCF-7, HeLa, SH-SY5Y, COLO 320)

    • Compare band pattern and intensity against established patterns

  • Blocking peptide validation:

    • Pre-incubate antibody with immunizing peptide before Western blot

    • True RPL37A bands should disappear or significantly diminish

  • siRNA knockdown controls:

    • Transfect cells with RPL37A-specific siRNA

    • Compare with control siRNA (e.g., siLuc)

    • RPL37A-specific bands should decrease in intensity proportional to knockdown efficiency

  • Loading control normalization:

    • Use housekeeping proteins distinct from ribosomal machinery (e.g., GAPDH, β-actin)

    • This helps distinguish between specific RPL37A changes and global ribosomal alterations

  • Cross-antibody validation:

    • When possible, compare results using antibodies targeting different RPL37A epitopes

    • Consistent patterns across different antibodies increase confidence in specificity

When unexpected bands appear, consider potential splice variants, proteolytic fragments, or post-translational modifications that may affect mobility.

What are the common causes of variability in RPL37A antibody performance between experiments?

Variability in RPL37A antibody performance can stem from several sources:

  • Antibody storage and handling:

    • Repeated freeze-thaw cycles degrade performance

    • Improper temperature storage accelerates antibody deterioration

    • Bacterial contamination from improper handling

  • Sample preparation variations:

    • Lysis buffer composition affects epitope accessibility

    • Incomplete denaturation for SDS-PAGE applications

    • Protein degradation during extraction

    • Phosphatase or protease inhibitor inconsistencies

  • Technical variations:

    • Inconsistent transfer efficiency in Western blotting

    • Variable blocking efficiency between experiments

    • Secondary antibody batch differences

    • Incubation time and temperature fluctuations

  • Biological variations:

    • Cell culture density and passage number affects RPL37A expression

    • Serum lot variations can influence basal expression

    • Mycoplasma contamination alters cellular physiology

  • Detection system variables:

    • ECL reagent aging

    • Imager sensitivity settings

    • Development timing inconsistencies

  • Cell treatment conditions:

    • RPL37A expression changes with cellular stress

    • Serum starvation conditions vary between labs

    • Confluency at harvest affects expression levels

To minimize variability:

  • Standardize protocols with detailed documentation

  • Prepare larger antibody aliquots to reduce freeze-thaw cycles

  • Include consistent positive controls in each experiment

  • Maintain detailed records of reagent lots and experimental conditions

How should researchers interpret discrepancies between RPL37A protein detection and mRNA expression data?

When encountering discrepancies between RPL37A protein and mRNA levels:

  • Consider post-transcriptional regulation:

    • RPL37A is subject to complex regulatory mechanisms beyond transcription

    • As a ribosomal protein, its translation may be autoregulated in response to ribosome assembly status

  • Protein stability factors:

    • RPL37A protein may have different half-life under various cellular conditions

    • Proteasomal degradation rates can vary independently of mRNA levels

    • Stress conditions can selectively affect protein stability without immediate mRNA changes

  • Methodological considerations:

    • Primer specificity in qPCR may detect splice variants differently than antibodies

    • Antibody epitopes may be masked by protein interactions or modifications

    • Detection sensitivity differences between protein and RNA methods

  • Temporal dynamics:

    • Time-course analysis may reveal delays between mRNA changes and protein accumulation

    • RPL37A mRNA and protein turnover rates may differ substantially

  • Cellular compartmentalization:

    • RPL37A protein may redistribute between cellular compartments under certain conditions

    • Some extraction methods may not efficiently recover all cellular pools

  • Experimental approach for resolution:

    • Perform time-course studies to detect temporal relationships

    • Use protein synthesis inhibitors (cycloheximide) to determine protein half-life

    • Employ proteasome inhibitors to assess degradation contribution

    • Consider ribosome profiling to assess translational efficiency

How can RPL37A antibodies be effectively utilized to study its role in the Mdm2-p53 pathway?

To investigate RPL37A's role in the Mdm2-p53 pathway:

  • Co-immunoprecipitation studies:

    • Use RPL37A antibody to immunoprecipitate endogenous protein complexes

    • Analyze for presence of Mdm2 and p53 by Western blot

    • Validated in H1299 (p53-null) and U2OS (p53-containing) cell lines

    • Optimal amount: 0.5-4.0 μg antibody per 1.0-3.0 mg total protein lysate

  • Pathway activation analysis:

    • Overexpress RPL37A and monitor p53 stabilization by Western blot

    • Assess downstream targets (p21, Puma) by qRT-PCR and Western blot

    • Analyze p53 post-translational modifications (phosphorylation, acetylation)

    • Note that RPL37A specifically upregulates p21 and Puma but not Mdm2, Noxa or Bax

  • Cell cycle analysis:

    • Transfect cells with tagged RPL37A

    • Perform flow cytometry to quantify cell cycle distribution

    • RPL37A expression increases sub-G1 phase (apoptosis) and arrests cells in G2 phase rather than G1

    • Compare with effects of other ribosomal proteins (e.g., RPS15, RPS20)

  • p53-independent effects:

    • Use p53-null cell lines (H1299) to identify p53-independent functions

    • RPL37A can interact with Mdm2 even in the absence of p53

  • MdmX regulation studies:

    • RPL37A reduces MdmX mRNA and protein levels without direct protein interaction

    • Use qRT-PCR to monitor MdmX mRNA changes following RPL37A overexpression

    • This mechanism differs from RPS15 and RPS20, which physically interact with MdmX

These approaches should be integrated to build a comprehensive understanding of RPL37A's specific role in the Mdm2-p53 regulatory network.

What methodological approaches can be used to validate RPL37A as a biomarker in cancer tissue samples?

Validating RPL37A as a cancer biomarker requires a multi-faceted approach:

  • Immunohistochemistry optimization:

    • Establish scoring criteria for RPL37A expression (high vs. low)

    • Use tissue microarrays for consistent processing of multiple samples

    • Implement digital pathology for quantitative assessment

    • Determine optimal cutoff values using receiver operating characteristic (ROC) curves

  • Correlation with clinical outcomes:

    • Analyze RPL37A expression in relation to:

      • Response to neoadjuvant chemotherapy (PCR rate)

      • Disease-free survival (DFS)

      • Cancer-specific survival (CSS)

    • Use multivariate Cox regression models to assess independent prognostic value

  • Comparative biomarker assessment:

    • Compare RPL37A with established markers (ER, PR, HER2, Ki67)

    • Evaluate RPL37A in combination with other potential biomarkers (MTSS1, HTRA1)

    • Determine if RPL37A adds independent value to existing prognostic models

  • Mechanism investigation:

    • Correlate RPL37A expression with cellular features (proliferation, necrosis, inflammatory infiltrate)

    • Assess relationship with molecular subtypes of cancer

    • Investigate connection to drug resistance mechanisms

  • Validation cohorts:

    • Confirm findings in independent patient cohorts

    • Include diverse ethnic backgrounds and treatment protocols

    • Validate in prospective studies when possible

  • Multi-omic integration:

    • Correlate protein expression with genomic and transcriptomic data

    • Evaluate RPL37A gene alterations (mutation, amplification) in relation to protein expression

    • Consider epigenetic regulation of RPL37A expression

Research has shown RPL37A is particularly valuable as a biomarker for response to neoadjuvant chemotherapy and for prognosis in breast cancer , with high expression associated with increased likelihood of pathological complete response.

How can researchers design experiments to distinguish between RPL37A's canonical ribosomal functions and its non-canonical roles in cell signaling?

Differentiating between RPL37A's canonical and non-canonical functions requires sophisticated experimental design:

  • Structure-function analysis:

    • Generate RPL37A mutants that selectively disrupt:

      • Ribosome incorporation (using ribosome structure data)

      • Mdm2 binding domains (based on interaction mapping)

    • Assess each mutant's ability to:

      • Incorporate into ribosomes (by sucrose gradient fractionation)

      • Bind Mdm2 (by co-immunoprecipitation)

      • Activate p53 (by reporter assays)

      • Affect cell cycle (by flow cytometry)

  • Cellular compartmentalization studies:

    • Use subcellular fractionation to separate:

      • Nucleolar (ribosome assembly)

      • Cytoplasmic (mature ribosomes)

      • Free protein pools (potentially signaling-related)

    • Immunofluorescence with confocal microscopy to track RPL37A localization under:

      • Normal conditions

      • Ribosomal stress (actinomycin D treatment)

      • DNA damage (radiation or chemotherapeutic agents)

  • Temporal dynamics:

    • Implement RPL37A inducible expression systems

    • Monitor time-course of:

      • Ribosome incorporation

      • p53 pathway activation

      • Cell cycle changes

      • Global translation rates

    • Early events may indicate direct signaling, while delayed events may reflect ribosomal functions

  • Ribosome-specific depletion:

    • Design siRNAs targeting UTRs specific to RPL37A mRNA

    • Compare with coding region siRNAs

    • Assess differential effects on ribosome assembly versus signaling

  • Separation of functions through interactor analysis:

    • Identify proteins uniquely interacting with free versus ribosome-incorporated RPL37A

    • Use BioID or APEX proximity labeling in different cellular compartments

    • Validate key interactors through reciprocal co-immunoprecipitation

  • Translational impact analysis:

    • Perform ribosome profiling after RPL37A manipulation

    • Identify specific mRNAs affected by RPL37A depletion

    • Determine if p53 pathway effects are secondary to translational changes

These approaches collectively help distinguish RPL37A's direct signaling roles from indirect effects of altered ribosome function .

What considerations are important when using RPL37A antibodies to study its involvement in stress response pathways?

When investigating RPL37A in stress response pathways:

  • Stress-specific modifications:

    • Consider that RPL37A may undergo post-translational modifications during stress

    • Use phospho-specific or modification-specific antibodies if available

    • Perform 2D gel electrophoresis to resolve modified forms

  • Dynamic expression changes:

    • Implement careful time-course experiments

      • Include early time points (15, 30, 60 minutes)

      • Extend to later phases (6, 12, 24 hours)

    • Monitor both protein levels and subcellular distribution

    • Different stressors may elicit distinct temporal patterns

  • Stress-specific localization:

    • Track RPL37A movement between:

      • Nucleolus (site of ribosome assembly)

      • Nucleoplasm (potential site for Mdm2 interaction)

      • Cytoplasm (site of mature ribosomes)

    • Use subcellular fractionation complemented by immunofluorescence

  • Stress type specificity:

    • Compare RPL37A response across multiple stressors:

      • Ribosomal stress (actinomycin D, 5-FU)

      • DNA damage (radiation, doxorubicin)

      • ER stress (thapsigargin, tunicamycin)

      • Nutrient deprivation (amino acid starvation, glucose limitation)

    • Determine if RPL37A behaves as a specific or general stress sensor

  • Stoichiometry considerations:

    • Assess relative levels of free versus ribosome-incorporated RPL37A

    • Determine if stress alters this distribution

    • Consider how detection methods might be biased toward specific pools

  • Cross-regulatory effects:

    • Examine how RPL37A interacts with other ribosomal proteins during stress

    • Determine if it functions independently or cooperatively with proteins like RPL11

    • Investigate whether it behaves as a "detector" or "effector" RP under different conditions

  • Translation consequences:

    • Measure global translation rates (puromycin incorporation)

    • Assess polysome profiles

    • Determine if RPL37A-mediated signaling occurs before or after translation inhibition

Research indicates RPL37A shows both "detector" and "effector" properties in stress responses, with distinct mechanisms from other ribosomal proteins like RPL11 .

How does RPL37A antibody performance compare with antibodies against other ribosomal proteins in multiplex studies?

When incorporating RPL37A antibodies into multiplex studies with other ribosomal protein antibodies:

  • Epitope compatibility:

    • RPL37A antibodies target various epitopes, commonly within regions 21-70

    • Ensure epitopes of multiplexed antibodies are sufficiently distinct to prevent competitive binding

    • Consider how ribosome structure may limit accessibility of certain epitopes

  • Species cross-reactivity differences:

    • RPL37A antibodies consistently detect human, mouse, and rat proteins

    • Other RP antibodies may have different species profiles

    • In cross-species studies, verify all antibodies work consistently across target species

  • Detection sensitivity variations:

    • RPL37A baseline expression varies across tissues and cell types

    • Standardize detection methods to accommodate different abundance levels

    • Consider sequential detection strategies for widely different expression levels

  • Size discrimination considerations:

    • RPL37A typically appears at 10 kDa

    • For multiplexing with similarly sized RPs (e.g., RPS20 at ~13 kDa):

      • Use higher resolution gels (12-15% acrylamide)

      • Consider labeled primary antibodies for simultaneous detection

      • Employ sequential stripping and reprobing with verification of complete stripping

  • Functional context integration:

    • When studying p53 pathway components:

      • RPL37A functions differently from RPS15 and RPS20 in MdmX regulation

      • RPL37A specifically upregulates p21 and Puma but not other p53 targets

    • Design multiplex panels that capture these functional distinctions

  • Isotype and host considerations:

    • Most RPL37A antibodies are rabbit polyclonal

    • For co-detection, combine with antibodies from different host species

    • Consider monoclonal alternatives for long-term reproducibility

This integrated approach allows researchers to leverage the distinct properties of RPL37A alongside other ribosomal proteins in complex experimental designs.

What methodological strategies can resolve contradictory findings in RPL37A research regarding its cell cycle effects?

To resolve contradictions in RPL37A cell cycle research:

  • Expression system differences:

    • Transient overexpression of RPL37A induces G2 arrest

    • Stable GFP-RPL37A expression reportedly causes G1 arrest

    • Reconciliation approaches:

      • Compare expression levels between systems

      • Assess tag interference with function

      • Examine adaptation in stable lines versus acute response

  • Cell type dependencies:

    • Systematically test RPL37A effects across:

      • p53 wild-type versus null backgrounds

      • Primary versus transformed cells

      • Different tissue origins

    • Document cell type-specific responses with consistent methodology

  • Temporal dynamics:

    • Implement time-course experiments:

      • Early (6-24h): May show G2 predominance

      • Late (48-72h): May shift to G1 arrest

    • Use synchronized cell populations to detect phase-specific effects

  • Dose-dependency analysis:

    • Titrate RPL37A expression levels using:

      • Inducible expression systems

      • Different transfection amounts

    • Determine if cell cycle effects show threshold or linear relationships

  • Context-dependent signaling:

    • Assess RPL37A effects under:

      • Normal growth conditions

      • Nutrient limitation

      • DNA damage

      • Ribosomal stress

    • Determine if context alters the direction of cell cycle effects

  • Mechanistic resolution through pathway analysis:

    • Compare p53-dependent versus independent mechanisms

    • Evaluate p21 induction timing relative to cell cycle changes

    • Assess differential regulation of cyclins and CDKs

    • Examine activation of checkpoint kinases (Chk1/2)

  • Technical considerations:

    • Standardize cell cycle analysis methods:

      • Flow cytometry parameters

      • DNA content versus BrdU incorporation

      • Cell synchronization approaches

    • Include appropriate controls (e.g., known G1 and G2 arrestors)

Research indicates that RPL37A can specifically induce G2 arrest while activating p21, which typically mediates G1 arrest . This apparent contradiction may reflect complex pathway crosstalk that requires careful dissection.

How should researchers approach the integration of RPL37A antibody data with multi-omic datasets in cancer research?

Integration of RPL37A antibody data with multi-omic cancer datasets requires systematic approaches:

  • Cross-platform normalization:

    • Develop standardized RPL37A expression scores across:

      • Immunohistochemistry (H-score or percent positive cells)

      • Proteomics (spectral counts or intensity-based metrics)

      • Transcriptomics (RNA-Seq or microarray data)

    • Use reference samples processed across all platforms

  • Correlation analysis frameworks:

    • Assess relationships between:

      • RPL37A protein levels (antibody-based)

      • mRNA expression (transcriptomics)

      • Gene copy number (genomics)

      • Promoter methylation (epigenomics)

    • Identify discordant samples for mechanistic investigation

  • Pathway context integration:

    • Map RPL37A within functional networks:

      • Ribosome biogenesis

      • p53 regulatory network

      • Cell cycle control

      • Treatment response pathways

    • Use protein-protein interaction data to build extended networks

  • Clinical outcome correlation:

    • Stratify patients by RPL37A protein expression

    • Integrate with molecular subtypes

    • Develop composite biomarkers incorporating RPL37A with other markers

    • Validate in independent cohorts with multivariate analysis

  • Treatment response prediction:

    • Correlate RPL37A levels with:

      • Neoadjuvant chemotherapy response

      • Pathological complete response rates

      • Disease-free and cancer-specific survival

    • Identify molecular features that modify RPL37A's predictive value

  • Technical considerations:

    • Document antibody clone, dilution, and scoring method

    • Include measurement uncertainty in integrative models

    • Consider tissue heterogeneity in bulk measurements

    • When possible, use spatial transcriptomics to correlate with IHC

  • Computational approaches:

    • Employ multivariate dimensionality reduction

    • Consider Bayesian integration frameworks

    • Use machine learning to identify complex patterns

    • Validate with independent datasets

Research has established RPL37A as a potential biomarker for response to neoadjuvant chemotherapy in breast cancer , and integrative approaches can further refine its clinical utility by placing it in broader molecular context.

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.