MRPS26 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
The antibody is supplied in phosphate buffered saline (PBS) with 0.1% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery timelines.
Synonyms
MRPS26 antibody; C20orf193 antibody; RPMS13 antibody; 28S ribosomal protein S26 antibody; mitochondrial antibody; MRP-S26 antibody; S26mt antibody; 28S ribosomal protein S13 antibody; mitochondrial antibody; MRP-S13 antibody; S13mt antibody; Mitochondrial small ribosomal subunit protein mS26 antibody
Target Names
MRPS26
Uniprot No.

Target Background

Database Links

HGNC: 14045

OMIM: 611988

KEGG: hsa:64949

STRING: 9606.ENSP00000369682

UniGene: Hs.18946

Protein Families
Mitochondrion-specific ribosomal protein mS26 family
Subcellular Location
Mitochondrion.

Q&A

How do I select the most appropriate MRPS26 antibody for my research application?

Selection should be based on:

  • Application compatibility: Verify validated applications for each antibody (WB, IHC, IP, ELISA, ICC, IF) and choose one specifically validated for your intended application .

  • Species reactivity: Confirm reactivity with your experimental model. Some antibodies show verified reactivity with human, mouse, and rat samples, while others may have broader or more limited species reactivity .

  • Antibody format: Consider whether an unconjugated antibody or one conjugated to a reporter molecule (e.g., HRP) better suits your experimental design .

  • Epitope recognition: Different antibodies target different regions (N-terminal, internal, C-terminal). For domain-specific studies, choose an antibody targeting the relevant region .

  • Validation evidence: Review validation data from manufacturers, including positive controls and reference tissues/cells shown to express the target .

ApplicationRecommended Dilution RangeExample Positive Controls
Western Blot1:500-1:2000HepG2 cells, HeLa cells, mouse brain tissue
Immunohistochemistry1:20-1:200Human skin cancer tissue, human liver cancer tissue
Immunoprecipitation0.5-4.0 μg per 1-3 mg proteinHepG2 cells
ELISA1:20000-1:80000Varies by kit

What is the difference between polyclonal and monoclonal MRPS26 antibodies, and how does this impact experimental design?

The search results indicate that most commercially available MRPS26 antibodies are polyclonal, derived from rabbits . This distinction has significant implications for research:

Polyclonal MRPS26 antibodies (such as those in the search results):

  • Recognize multiple epitopes within the target protein, potentially increasing sensitivity for detecting low abundance targets

  • May provide more robust detection when protein conformation or post-translational modifications are variable

  • Show batch-to-batch variation that may necessitate standardization between experiments

  • Are particularly useful for applications like immunoprecipitation where binding to multiple epitopes enhances capture efficiency

Monoclonal antibodies (though not specifically mentioned in search results for MRPS26):

  • Recognize a single epitope with high specificity

  • Provide consistent performance with minimal batch-to-batch variation

  • May be less sensitive to conformational changes in the target protein

  • Are ideal for applications requiring high reproducibility across experiments

For challenging applications like detecting native protein complexes, researchers should consider using polyclonal antibodies that can recognize the protein in its native conformation, while applications requiring extreme specificity might benefit from monoclonal antibodies if available .

What are the optimal conditions for using MRPS26 antibodies in Western blot applications?

Based on the available data, optimal Western blotting conditions for MRPS26 antibodies include:

  • Sample preparation: Most validated in cell lysates (HepG2, HeLa) and tissue homogenates (mouse brain) . For mitochondrial enrichment, consider subcellular fractionation prior to Western blotting.

  • Antibody dilutions: Use within the recommended range of 1:500-1:2000 . Start with a mid-range dilution (1:1000) and adjust based on signal-to-noise ratio.

  • Protein loading: Load 20-30 μg of total protein per lane for whole cell lysates. For purified mitochondrial fractions, 10-15 μg may be sufficient.

  • Expected molecular weight: Look for a band at approximately 24 kDa, which corresponds to the observed molecular weight of MRPS26 .

  • Blocking conditions: Use 5% non-fat dry milk or BSA in TBST. For phospho-specific detection, BSA is preferred over milk.

  • Incubation times: Primary antibody incubation can be performed overnight at 4°C or for 2 hours at room temperature for optimal binding.

  • Controls: Include positive controls (e.g., HepG2 or HeLa cell lysates) and consider including a loading control for normalization .

  • Detection methods: Both chemiluminescence and fluorescence-based detection systems are compatible, with HRP-conjugated secondary antibodies being commonly used .

Importantly, MRPS26 antibodies have been validated in multiple publications, confirming their specificity and reliability for Western blot applications .

How should I optimize immunohistochemistry protocols using MRPS26 antibodies?

For optimal IHC results with MRPS26 antibodies:

  • Antigen retrieval: Use TE buffer pH 9.0 as the preferred method. Alternatively, citrate buffer pH 6.0 may be used, though potentially with different efficacy .

  • Antibody dilution: Start with a dilution range of 1:20-1:200, titrating to determine optimal concentration for your specific tissue type .

  • Positive control tissues: Include human skin cancer tissue or human liver cancer tissue as positive controls, as these have been validated to express detectable levels of MRPS26 .

  • Incubation conditions: Incubate sections with primary antibody overnight at 4°C in a humidified chamber to maximize specific binding while minimizing background.

  • Detection systems: Both ABC (Avidin-Biotin Complex) and polymer-based detection systems are compatible. For low expression targets, consider using amplification systems.

  • Counterstaining: Use hematoxylin for nuclear counterstaining, but avoid overstaining which may obscure specific MRPS26 signals.

  • Fixation considerations: MRPS26 antibodies have been validated with formalin-fixed, paraffin-embedded (FFPE) tissues, but fixation time should be optimized to preserve epitope accessibility .

  • Multi-labeling experiments: For co-localization studies with other mitochondrial markers, sequential immunostaining is recommended over simultaneous protocols to minimize cross-reactivity.

What methodology should I follow for immunoprecipitation experiments using MRPS26 antibodies?

Based on validated protocols, the following methodology is recommended for MRPS26 immunoprecipitation:

  • Antibody amount: Use 0.5-4.0 μg of MRPS26 antibody per 1.0-3.0 mg of total protein lysate .

  • Lysis conditions: Use a buffer containing 150 mM NaCl, 1% NP-40 or Triton X-100, 50 mM Tris pH 8.0, and protease inhibitors. For mitochondrial protein complexes, milder detergents like digitonin (0.5-1%) may better preserve protein-protein interactions.

  • Pre-clearing step: Pre-clear lysates with protein A/G beads to reduce non-specific binding.

  • Antibody binding: Incubate clarified lysate with MRPS26 antibody overnight at 4°C with gentle rotation.

  • Immunoprecipitation: Add protein A/G beads and incubate for 1-4 hours at 4°C.

  • Washing conditions: Perform 3-5 washes with lysis buffer containing reduced detergent concentration to minimize background while preserving specific interactions.

  • Elution methods: For subsequent mass spectrometry analysis, consider on-bead digestion or mild elution conditions to preserve protein integrity.

  • Controls: Include an isotype control (rabbit IgG) processed identically to assess non-specific binding .

  • Validated samples: HepG2 cells have been confirmed as positive controls for MRPS26 immunoprecipitation .

For studying MRPS26 in the context of mitochondrial ribosome complexes, crosslinking before immunoprecipitation may help capture transient interactions with other ribosomal components.

How can I troubleshoot weak or absent MRPS26 signal in Western blot experiments?

When experiencing weak or absent signals when using MRPS26 antibodies in Western blot applications, consider the following troubleshooting approaches:

  • Sample preparation issues:

    • Ensure proper cell lysis and protein extraction using buffers containing appropriate detergents

    • Check protein concentration measurement methodology

    • Verify sample integrity by examining housekeeping proteins

    • Consider mitochondrial enrichment for low-abundance samples

  • Antibody-related factors:

    • Verify antibody concentration (try higher concentration within recommended range: 1:500 instead of 1:2000)

    • Check antibody storage conditions (-20°C is recommended for most MRPS26 antibodies)

    • Avoid repeated freeze-thaw cycles that may reduce antibody activity

    • Consider using fresh antibody aliquots if old stocks show diminished activity

    • Verify recognition of the specific species being tested (human, mouse, rat, etc.)

  • Protocol modifications:

    • Extend primary antibody incubation time (overnight at 4°C)

    • Optimize blocking conditions (5% BSA may reduce background compared to milk for some applications)

    • Try alternative membrane types (PVDF vs. nitrocellulose)

    • Increase protein loading (up to 50 μg per lane)

    • Enhance signal using more sensitive detection reagents

  • Epitope accessibility:

    • If using reducing conditions, try non-reducing conditions or vice versa

    • Adjust SDS concentration in sample buffer

    • Consider native gel electrophoresis if the epitope is conformationally sensitive

  • Positive controls:

    • Include validated positive controls such as HepG2 cells, HeLa cells, or mouse brain tissue known to express MRPS26

What are the common sources of non-specific binding when using MRPS26 antibodies, and how can they be minimized?

Non-specific binding can compromise data interpretation with MRPS26 antibodies. Key sources and mitigation strategies include:

  • Cross-reactivity with related proteins:

    • Verify the antibody has been validated against the specific protein target

    • Consider using antibodies targeting different epitopes of MRPS26 to confirm specificity

    • For critical experiments, validate results with alternative methods like mass spectrometry

  • Insufficient blocking:

    • Optimize blocking conditions (5% BSA or milk in TBST)

    • Extend blocking time to 1-2 hours at room temperature

    • Add 0.1-0.3% Tween-20 to washing buffers to reduce hydrophobic interactions

  • Secondary antibody issues:

    • Use highly cross-adsorbed secondary antibodies

    • Dilute secondary antibodies appropriately (typically 1:5000-1:10000)

    • Pre-adsorb secondary antibodies with tissue/cell lysates from the experimental species

  • Sample preparation:

    • Ensure complete lysis and denaturation of proteins for SDS-PAGE

    • Pre-clear samples by centrifugation to remove insoluble material

    • For IP applications, pre-clear with protein A/G beads before adding the specific antibody

  • Fixation artifacts in IHC/ICC:

    • Optimize fixation time and conditions

    • Perform proper antigen retrieval (TE buffer pH 9.0 recommended for MRPS26)

    • Include appropriate negative controls (omitting primary antibody, using non-immune IgG)

  • Endogenous biotin or peroxidase activity:

    • Block endogenous peroxidase with hydrogen peroxide treatment before antibody application

    • For biotin-based detection systems, block endogenous biotin using avidin/biotin blocking kits

By implementing these strategies, researchers can minimize non-specific signals and improve the reliability of MRPS26 detection across experimental platforms.

How do storage conditions and handling affect MRPS26 antibody performance?

Optimal storage and handling are critical for maintaining MRPS26 antibody performance:

  • Temperature considerations:

    • Store unconjugated antibodies at -20°C for long-term storage

    • HRP-conjugated antibodies may require 4°C storage in the dark to preserve enzymatic activity

    • Avoid repeated freeze-thaw cycles by preparing small aliquots upon receipt

  • Buffer composition effects:

    • Most MRPS26 antibodies are supplied in PBS with stabilizers like glycerol (50%) and preservatives like sodium azide (0.02%)

    • Some formulations include BSA (0.1-1%) for additional stability

    • Do not dilute stock solutions unless preparing working aliquots

  • Reconstitution practices (for lyophilized formats):

    • Reconstitute in 100 μl of sterile distilled water with 50% glycerol as recommended

    • Allow complete dissolution before use (gentle mixing, no vortexing)

    • Prepare at the recommended concentration (typically 1 mg/ml after reconstitution)

  • Working dilution stability:

    • Diluted antibody solutions are less stable than stock concentrations

    • For diluted working solutions, store at 4°C and use within 1-2 weeks

    • Add 0.02% sodium azide to working dilutions to prevent microbial growth

  • Shipment considerations:

    • Verify antibody activity after shipment, especially if shipping conditions were suboptimal

    • Allow refrigerated antibodies to equilibrate to room temperature before opening to prevent condensation

  • Contamination prevention:

    • Use sterile technique when handling antibody solutions

    • Avoid introducing bacteria or fungi that could degrade the antibody or produce proteases

Following these guidelines will help maintain antibody performance and ensure reproducible results across experiments.

How can MRPS26 antibodies be used to investigate mitochondrial ribosome assembly and function?

MRPS26 antibodies offer several approaches for investigating mitochondrial ribosome assembly and function:

  • Co-immunoprecipitation studies: Use MRPS26 antibodies (0.5-4.0 μg per 1-3 mg lysate) to pull down the small mitochondrial ribosomal subunit and associated proteins . This approach can:

    • Identify novel interaction partners through mass spectrometry

    • Confirm known interactions with other mitochondrial ribosomal proteins

    • Investigate how pathogenic mutations affect ribosome assembly

  • Proximity labeling methods: Combine MRPS26 antibodies with proximity labeling techniques (BioID, APEX) to map the spatial organization of mitochondrial ribosomes:

    • Immunolocalize MRPS26 after proximity labeling to validate specific ribosomal neighborhoods

    • Use MRPS26 antibodies to verify successful pull-down of labeled complexes

  • Ribosome profiling validation: MRPS26 antibodies can verify the integrity of mitochondrial ribosomes isolated for ribosome profiling:

    • Confirm the presence of MRPS26 in gradient fractions containing assembled small subunits

    • Monitor MRPS26 incorporation into ribosomes under different cellular conditions

  • Structural studies integration: When performing cryo-EM studies of mitochondrial ribosomes:

    • Use MRPS26 antibodies for Western blot verification of samples prior to structural analysis

    • Validate protein identification in resolved structures through immunogold labeling

  • Mitochondrial translation assays: MRPS26 antibodies can correlate ribosome assembly with translation activity:

    • Monitor MRPS26 levels in parallel with mitochondrial translation efficiency

    • Assess how depletion of other ribosomal components affects MRPS26 incorporation

  • Tissue-specific expression patterns: Using IHC applications (dilutions 1:20-1:200), investigate tissue-specific variations in MRPS26 expression that may reflect differing mitochondrial translation requirements .

These approaches provide complementary perspectives on how MRPS26 contributes to mitochondrial ribosome formation and function across different experimental systems.

What approaches can be used to study the relationship between MRPS26 and mitochondrial disease models?

Several methodological approaches can be employed to investigate MRPS26's role in mitochondrial disease:

  • Comparative expression analysis:

    • Use validated MRPS26 antibodies in Western blot (1:500-1:2000) to compare expression levels between healthy and disease tissues/cells

    • Perform quantitative immunohistochemistry (1:20-1:200) on patient-derived tissues versus controls

    • Correlate MRPS26 levels with clinical parameters or disease progression markers

  • Post-translational modification studies:

    • Investigate disease-associated PTMs of MRPS26 using specialized antibodies

    • Combine immunoprecipitation with mass spectrometry to identify modified residues

    • Compare PTM patterns between normal and pathological states

  • Protein-protein interaction networks:

    • Use MRPS26 antibodies for co-immunoprecipitation (0.5-4.0 μg per 1-3 mg lysate) followed by mass spectrometry to map interaction partners

    • Compare interaction profiles between healthy and diseased states

    • Identify disease-specific interactions that may represent therapeutic targets

  • Functional knockdown/knockout validation:

    • Correlate MRPS26 depletion phenotypes with disease manifestations

    • Use MRPS26 antibodies to confirm knockdown efficiency and investigate compensatory mechanisms

    • Rescue experiments with wild-type vs. mutant MRPS26 variants

  • Mitochondrial translation activity:

    • Measure translation of mitochondrially-encoded proteins in disease models

    • Correlate translation defects with MRPS26 abundance, localization, or incorporation into ribosomes

    • Investigate tissue-specific translation defects using immunohistochemistry in disease tissues

  • Patient-derived cell models:

    • Apply MRPS26 antibodies to characterize patient-derived fibroblasts, induced pluripotent stem cells, or differentiated cell types

    • Compare mitochondrial ribosome assembly between patient and control cells

    • Test potential therapeutic compounds for normalization of MRPS26-related defects

These approaches can provide mechanistic insights into how MRPS26 dysfunction contributes to mitochondrial disease pathogenesis and identify potential therapeutic strategies.

How can MRPS26 antibodies be integrated into multi-omics approaches for mitochondrial research?

Integration of MRPS26 antibodies into multi-omics research strategies enables comprehensive analysis of mitochondrial function:

These integrated approaches provide a comprehensive understanding of how MRPS26 contributes to mitochondrial function within the broader cellular context.

How should researchers quantify and normalize MRPS26 expression data from Western blot experiments?

  • Image acquisition considerations:

    • Capture images within the linear dynamic range of the detection system

    • Avoid saturated pixels that compromise quantification accuracy

    • Use the same exposure settings for experimental and control samples

    • For fluorescence-based detection, minimize photobleaching

  • Band intensity measurement:

    • Use scientific image analysis software (ImageJ, ImageLab, etc.)

    • Define consistent regions of interest for all samples

    • Subtract local background from each measurement

    • Analyze the 24 kDa band corresponding to MRPS26's observed molecular weight

  • Normalization strategies:

    • Loading controls: Normalize to housekeeping proteins (β-actin, GAPDH) for whole cell lysates

    • Mitochondrial controls: For mitochondrial enriched samples, normalize to stable mitochondrial proteins (VDAC, COX IV) rather than cellular housekeeping genes

    • Total protein normalization: Consider Ponceau S or SYPRO Ruby staining as alternatives to individual loading controls

  • Statistical analysis:

    • Perform experiments with at least three biological replicates

    • Apply appropriate statistical tests based on data distribution

    • Present both individual data points and means with error bars

    • Consider normality testing before applying parametric statistics

  • Data presentation best practices:

    • Include representative blot images showing MRPS26 and loading controls

    • Present quantification in graphical format with statistical significance indicated

    • Maintain consistent y-axis scaling when comparing across experiments

    • Indicate antibody dilution used (within 1:500-1:2000 range)

  • Validation approaches:

    • Confirm key findings with alternative MRPS26 antibodies targeting different epitopes

    • Correlate protein levels with functional outcomes

    • Consider complementary approaches (qPCR, proteomics) for comprehensive analysis

Following these guidelines ensures that MRPS26 expression data is accurately quantified, properly normalized, and statistically valid.

How can researchers interpret discrepancies in MRPS26 detection between different experimental techniques?

When facing discrepancies in MRPS26 detection across different experimental techniques, consider these analysis and resolution strategies:

  • Discrepancies between Western blot and immunohistochemistry:

    • Epitope accessibility: Fixation in IHC may mask epitopes that are accessible in denatured WB samples

    • Resolution approach: Try alternative antigen retrieval methods in IHC (TE buffer pH 9.0 is recommended)

    • Verification strategy: Use antibodies targeting different MRPS26 epitopes to confirm results

  • Differences between transcript and protein levels:

    • Biological explanation: Post-transcriptional regulation or protein stability differences

    • Resolution approach: Perform time-course experiments to detect temporal discrepancies

    • Verification strategy: Use protein synthesis or degradation inhibitors to assess MRPS26 turnover

  • Variations between antibodies:

    • Technical cause: Different epitope recognition, affinity, or specificity

    • Resolution approach: Validate with knockout/knockdown controls

    • Verification strategy: Compare results from multiple antibodies (e.g., C-terminal vs. internal epitope antibodies)

  • Cell type or tissue-specific discrepancies:

    • Biological explanation: Context-dependent expression or post-translational modifications

    • Resolution approach: Use positive control samples (HepG2, HeLa, mouse brain)

    • Verification strategy: Perform subcellular fractionation to assess localization differences

  • Methodology-dependent variations:

    • Technical cause: Buffer compatibility, detection sensitivity limits

    • Resolution approach: Optimize protocols for each specific application

    • Verification strategy: Use recombinant MRPS26 as a standard for calibration

  • Quantification discrepancies:

    • Technical cause: Different dynamic ranges between methods

    • Resolution approach: Establish standard curves for quantitative applications

    • Verification strategy: Use absolute quantification methods (e.g., AQUA peptides in mass spectrometry)

By systematically addressing these potential sources of discrepancy, researchers can reconcile apparently conflicting results and develop a more comprehensive understanding of MRPS26 biology.

What are the most rigorous controls to validate specificity when using MRPS26 antibodies in complex experimental systems?

To ensure maximum rigor when using MRPS26 antibodies, implement these critical controls:

  • Genetic knockout/knockdown validation:

    • Use CRISPR/Cas9 knockout cells or siRNA knockdown samples

    • Verify complete absence or significant reduction of signal in Western blot (1:500-1:2000)

    • Demonstrate restoration of signal with rescue constructs

    • Document degree of knockdown using qPCR in parallel with protein detection

  • Epitope blocking experiments:

    • Pre-incubate antibody with immunizing peptide or recombinant MRPS26

    • Demonstrate signal elimination in competitive blocking

    • Use titrated amounts of blocking peptide to establish specificity threshold

    • Include irrelevant peptides as negative controls for blocking

  • Multiple antibody validation:

    • Compare results using antibodies targeting different MRPS26 epitopes (internal regions vs. C-terminal)

    • Verify consistent patterns across polyclonal antibodies from different sources

    • Document any epitope-specific differences in detection efficiency

  • Orthogonal technique confirmation:

    • Correlate antibody-based detection with mass spectrometry identification

    • Compare results with alternative detection methods (e.g., GFP-tagged MRPS26)

    • Validate subcellular localization using fractionation followed by Western blot

  • Biologically relevant positive controls:

    • Include validated positive control samples (HepG2 cells, HeLa cells, mouse brain tissue)

    • Test tissues/cells known to have varying MRPS26 expression levels

    • Use recombinant MRPS26 as size and specificity reference

  • Methodological controls:

    • For IP experiments: include isotype control antibodies processed identically

    • For IHC/ICC: omit primary antibody while maintaining all other steps

    • For multiplexed detection: perform single-antibody controls to assess bleed-through

  • Cross-species validation:

    • Test antibody across species with high sequence homology to human MRPS26

    • Document species-specific detection patterns based on predicted reactivity

    • Correlate detection efficiency with sequence conservation in the target epitope

How might MRPS26 antibodies contribute to emerging research on mitochondrial dysfunction in neurodegenerative diseases?

MRPS26 antibodies offer significant potential for advancing understanding of mitochondrial contributions to neurodegeneration:

  • Characterization of disease-specific alterations:

    • Use Western blot (1:500-1:2000) to quantify MRPS26 levels in patient-derived tissues and cellular models

    • Apply immunohistochemistry (1:20-1:200) to map MRPS26 distribution in brain regions affected by neurodegeneration

    • Investigate correlations between MRPS26 levels and disease progression markers

  • Investigation of mitochondrial translation defects:

    • Employ MRPS26 antibodies to assess mitochondrial ribosome integrity in disease models

    • Correlate MRPS26 incorporation into ribosomes with translation efficiency of respiratory chain components

    • Compare post-translational modifications of MRPS26 between healthy and diseased states

  • Protein-protein interaction network analysis:

    • Use immunoprecipitation (0.5-4.0 μg per 1-3 mg lysate) coupled with mass spectrometry to identify disease-specific interactors

    • Investigate how pathogenic mutations in other proteins affect association with MRPS26

    • Map changes in the mitochondrial ribosome interactome across disease progression

  • Therapeutic target validation:

    • Monitor MRPS26 levels and incorporation into functional ribosomes following experimental therapies

    • Assess normalization of mitochondrial translation as a therapeutic endpoint

    • Develop high-throughput screens using MRPS26 antibodies to identify compounds that restore mitochondrial ribosome assembly

  • Biomarker development:

    • Evaluate MRPS26 or its modified forms as potential biomarkers for mitochondrial dysfunction

    • Correlate MRPS26 levels in accessible tissues with disease severity or progression

    • Develop sensitive immunoassays for quantification in clinical samples

  • Cell-type specific vulnerability assessment:

    • Use immunofluorescence co-labeling to investigate cell-type specific variations in MRPS26 expression

    • Determine whether vulnerable neuronal populations exhibit distinctive MRPS26 characteristics

    • Correlate MRPS26 expression patterns with regional sensitivity to neurodegeneration

These approaches could provide critical insights into mitochondrial contributions to neurodegenerative diseases and potentially identify novel therapeutic targets.

What methodological innovations might enhance the utility of MRPS26 antibodies for single-cell and spatial proteomics applications?

Emerging methodological advances can expand MRPS26 antibody applications in cutting-edge single-cell and spatial analysis:

  • Antibody conjugation strategies:

    • Direct conjugation to fluorophores with distinct spectral properties for multiplexed imaging

    • Conjugation to DNA barcodes for spatial transcriptomics-proteomics integration

    • Development of cleavable linkers for antibody-based proximity labeling

    • Creation of HRP or APEX2 conjugates for enhanced signal amplification in low-abundance settings

  • Microfluidic applications:

    • Integration with single-cell Western blot platforms

    • Adaptation for microfluidic immunocytochemistry with live-cell imaging

    • Development of automated microfluidic immunoprecipitation for minimal sample input

    • Optimization for circulating mitochondria analysis in liquid biopsies

  • High-content imaging innovations:

    • Super-resolution microscopy protocols optimized for MRPS26 detection

    • Integration with expansion microscopy for enhanced spatial resolution

    • Correlation with electron microscopy through CLEM (Correlative Light and Electron Microscopy)

    • Development of live-cell compatible antibody fragments for temporal analyses

  • Mass cytometry and imaging mass cytometry:

    • Metal-conjugated MRPS26 antibodies for CyTOF analysis

    • Integration into multiplexed ion beam imaging panels

    • Combination with mitochondrial functional probes for multiparametric analysis

    • Development of quantitative standards for absolute quantification

  • Spatial proteomics integration:

    • Validation protocols for MRPS26 detection in tissue sections with spatial transcriptomics

    • Co-detection workflows for MRPS26 with mitochondrially-encoded transcripts

    • Development of cyclic immunofluorescence panels incorporating MRPS26

    • Integration with laser capture microdissection for region-specific analysis

  • Single-molecule detection methods:

    • Adaptation for proximity ligation assays to detect MRPS26 interactions

    • Development of aptamer-antibody hybrid probes for enhanced sensitivity

    • Integration with digital counting methods for absolute quantification

    • Optimization for single-molecule pull-down applications

These methodological innovations would significantly expand the utility of MRPS26 antibodies for investigating mitochondrial biology at single-cell resolution and within spatial contexts.

How might MRPS26 antibodies contribute to understanding the crosstalk between mitochondrial translation and cellular stress responses?

MRPS26 antibodies can provide valuable insights into the bidirectional relationship between mitochondrial translation and cellular stress:

  • Stress-induced alterations in MRPS26:

    • Monitor MRPS26 levels using Western blot (1:500-1:2000) following various stressors (oxidative, ER, nutrient, hypoxic)

    • Assess post-translational modifications using immunoprecipitation coupled with mass spectrometry

    • Track subcellular redistribution during stress using immunofluorescence

    • Investigate stress-responsive regulation of MRPS26 incorporation into ribosomes

  • Mitochondrial integrated stress response (ISRmt) correlation:

    • Analyze MRPS26 association with mitochondrial ribosomes during ISRmt activation

    • Correlate MRPS26-containing ribosome assembly with selective translation of stress-responsive transcripts

    • Investigate how ATF4 and other ISRmt factors influence MRPS26 expression or localization

    • Assess MRPS26 modifications in relation to stress-induced mitochondrial biogenesis

  • Interaction with stress granules and processing bodies:

    • Use co-immunoprecipitation (0.5-4.0 μg per 1-3 mg lysate) to identify stress-dependent interactions

    • Investigate whether MRPS26 or MRPS26-containing complexes associate with cytosolic RNA granules

    • Assess coordination between cytosolic and mitochondrial translation systems under stress

  • Mitochondrial quality control pathways:

    • Analyze MRPS26 fate during mitophagy using immunofluorescence

    • Investigate correlation between ribosomal disassembly and MRPS26 degradation

    • Assess MRPS26 recycling during mitochondrial biogenesis following stress

    • Monitor MRPS26 in relation to mitochondrial unfolded protein response activation

  • Methodological approaches:

    • Develop pulse-chase protocols to track MRPS26 dynamics during stress responses

    • Combine with proximity labeling to map stress-induced changes in the MRPS26 interactome

    • Implement multi-color live imaging using fluorescently tagged MRPS26 validated with antibodies

    • Apply quantitative proteomics to measure stress-induced changes in MRPS26 abundance and modifications

These approaches could reveal how mitochondrial translation machinery responds to and influences cellular stress responses, potentially identifying novel therapeutic targets for diseases involving mitochondrial dysfunction.

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.