RPL26 antibodies are utilized in diverse experimental workflows:
Detection of RPL26 in cell lysates: Validates protein expression levels in human, mouse, and rat cells (e.g., HEK293, K562, Jurkat).
Post-translational modification analysis: Identifies UFMylation (mono- and di-UFMylated forms) or phosphorylation.
Subcellular localization: Maps RPL26 to the nucleolus, cytoplasm, or ER-associated ribosomes.
Co-localization studies: Examines interactions with nucleolin or ribosomal partners.
Protein-protein interaction studies: Identifies RPL26 binding partners (e.g., UFM1, eIF4E).
RNA-ChIP: Captures RPL26-bound RNAs (e.g., p53/p73 mRNA 3′UTRs).
Quantitative analysis: Measures RPL26 levels in serum or tissue sections.
RPL26 antibodies have enabled critical insights into its biological roles:
UFMylation target: RPL26 is the primary substrate for UFM1 conjugation, with lysines 132 and 134 as modification sites. Antibodies detect mono- and di-UFMylated forms, linking UFMylation to ER protein homeostasis .
Functional impact: Disruption of RPL26 UFMylation (via 5KR mutants) impairs ER-associated degradation (ERAD), as shown in U2OS cells .
p53/p73 Translation: RPL26 binds to the 3′UTR of p53/p73 mRNA, enhancing their translation. Antibodies confirm RPL26’s interaction with eIF4E and its role in stress-induced p53 induction .
Protein stability: RPL26 stabilizes p73 by inhibiting MDM2-mediated degradation, as demonstrated in CRISPRi knockdown models .
RPL26 (Ribosomal Protein L26) is a component of the 60S large ribosomal subunit, also known as large ribosomal subunit protein uL24 or DBA11. With a calculated molecular weight of 17 kDa (though observed at 17-22 kDa in experimental conditions), this protein participates in the assembly and function of ribosomes essential for protein synthesis. RPL26 localizes primarily to ribosomes but can also interact with specific mRNAs to regulate translation. It serves as a useful marker for identifying Hippocampal Gyrus CA4 Neurons in neuroanatomical studies . The protein is evolutionarily conserved, with orthologs found in various species including mouse, rat, fly, canine, porcine, and monkey models .
RPL26 antibodies provide essential tools for studying ribosome biogenesis, translation regulation, and stress responses. Their significance extends to multiple research domains: (1) cancer biology, where altered ribosome function affects cellular transformation; (2) neuroscience, where RPL26 serves as a neuronal marker; (3) developmental biology, examining translation regulation during differentiation; and (4) RNA biology, investigating ribosome-associated regulatory mechanisms. These antibodies enable visualization, quantification, and isolation of RPL26-containing complexes through various experimental approaches including western blotting, immunoprecipitation, and immunofluorescence microscopy .
Research-grade RPL26 antibodies are available in multiple formats with distinct characteristics suited to different experimental needs. The primary classifications include: (1) Polyclonal antibodies, like the rabbit anti-RPL26 (17619-1-AP), which recognize multiple epitopes providing high sensitivity but potentially variable batch-to-batch consistency ; (2) Monoclonal antibodies offering exceptional specificity for single epitopes; (3) Various host species options including rabbit, mouse, and goat; (4) Specialized conjugates including FITC, HRP, and biotin-labeled variants for direct detection; and (5) Application-optimized formulations validated for specific techniques including western blotting, immunohistochemistry, immunofluorescence, and co-immunoprecipitation . Multiple vendors offer over 165 RPL26 antibody products across 21 suppliers, allowing researchers to select products with appropriate validation data for their specific experimental systems .
RPL26 antibodies demonstrate utility across multiple experimental platforms with varying levels of validation. Western blotting represents the most extensively validated application, with antibodies like 17619-1-AP showing consistent detection in human cell lines including K-562, HeLa, HepG2, and Jurkat cells at dilutions of 1:500-1:1000 . Immunofluorescence microscopy provides spatial information on RPL26 distribution within cells, particularly in nucleolar regions and cytoplasmic ribosomes, with recommended dilutions of 1:50-1:500 for optimal signal-to-noise ratios . Co-immunoprecipitation applications enable studying protein-protein interactions involving RPL26, particularly in ribosome assembly dynamics. Immunohistochemistry allows tissue-level localization, though requires careful optimization of antigen retrieval methods. For multiplex studies, RPL26 antibodies can be incorporated into flow cytometry panels with appropriate conjugates. The selection of application should be guided by available validation data for specific antibody clones .
Prior to implementing RPL26 antibodies in pivotal experiments, researchers should conduct systematic validation to ensure specificity and performance. Essential validation steps include: (1) Western blot analysis comparing the antibody's reactivity pattern against predicted molecular weight (17-22 kDa for RPL26); (2) Testing on negative controls such as RPL26 knockout or knockdown samples, with publications demonstrating this approach for certain antibodies ; (3) Cross-species validation when working with non-human models, considering cited reactivity in human and mouse samples ; (4) Testing across multiple cell lines to confirm consistent band patterns, as demonstrated with K-562, HeLa, HepG2, and Jurkat cells for some antibodies ; (5) Comparing results from multiple RPL26 antibodies targeting different epitopes to confirm concordant patterns; and (6) Conducting peptide blocking experiments to verify epitope-specific binding. These validation strategies should be customized based on experimental design and documented in publications .
For optimal Western blot detection of RPL26, researchers should follow these methodological guidelines: (1) Sample preparation should include complete cell lysis using RIPA or similar buffers with protease inhibitors to prevent degradation; (2) Loading 20-40 μg of total protein per lane on 12-15% SDS-PAGE gels to adequately resolve this low molecular weight protein; (3) Using wet transfer systems with PVDF membranes, which typically provide better results for ribosomal proteins than nitrocellulose; (4) Blocking with 5% non-fat dry milk in TBST for 1 hour at room temperature; (5) Incubating with primary RPL26 antibody at 1:500-1:1000 dilution overnight at 4°C ; (6) Washing thoroughly with TBST buffer (4 × 5 minutes); (7) Incubating with appropriate HRP-conjugated secondary antibody; and (8) Developing using enhanced chemiluminescence with exposure optimization to prevent saturation. The expected molecular weight range is 17-22 kDa, though additional bands may appear due to post-translational modifications or degradation products .
Successful immunofluorescence staining with RPL26 antibodies requires attention to several technical parameters: (1) Fixation method significantly impacts epitope accessibility, with 4% paraformaldehyde for 15 minutes typically preserving RPL26 antigenicity; (2) Permeabilization requires careful optimization, with 0.2% Triton X-100 for 5-10 minutes generally sufficient for accessing nuclear and cytoplasmic RPL26; (3) Primary antibody concentration should be tested at a range of 1:50-1:500 to determine optimal signal-to-noise ratio ; (4) Incubation time and temperature affect staining intensity, with overnight incubation at 4°C often producing superior results compared to shorter room temperature protocols; (5) Counterstaining with DAPI helps visualize nuclear localization patterns; (6) Including positive control cells with known RPL26 expression (such as HeLa cells) validates staining protocols ; and (7) Z-stack acquisition may be necessary to fully capture the three-dimensional distribution of RPL26 throughout the nucleolus and cytoplasm. The expected pattern includes nucleolar enrichment with diffuse cytoplasmic staining representing assembled ribosomes .
Multiple technical and biological factors can influence RPL26 antibody performance across experimental systems: (1) Antibody source and clonality—polyclonal antibodies typically offer higher sensitivity but potentially lower specificity compared to monoclonals; (2) Sample preparation methods—inadequate fixation, improper lysis, or protein degradation can significantly alter epitope availability; (3) Species cross-reactivity limitations—while some antibodies recognize human and mouse RPL26, verification is essential when working with other species like rat or non-mammalian models; (4) Post-translational modifications—phosphorylation, ubiquitination, or other modifications may mask epitopes or alter antibody recognition; (5) Ribosome assembly state—RPL26 epitopes may be differentially accessible in free versus assembled ribosomes; (6) Batch-to-batch variability—particularly relevant for polyclonal antibodies; and (7) Storage conditions—repeated freeze-thaw cycles or improper temperature control can compromise antibody performance over time .
Non-specific binding represents a common challenge when working with ribosomal protein antibodies. Effective strategies to minimize this issue include: (1) Optimizing blocking conditions—testing alternative blocking agents such as BSA, casein, or commercial blocking solutions beyond standard non-fat milk; (2) Increasing wash stringency—adding up to 0.1% SDS or increasing salt concentration in wash buffers; (3) Titrating antibody concentration—testing serial dilutions beyond manufacturer recommendations to identify optimal signal-to-noise ratio; (4) Pre-absorbing antibodies—incubating with cell lysates from non-relevant species to remove cross-reactive components; (5) Using gradient SDS-PAGE gels (4-20%) to better resolve RPL26 from potentially cross-reactive proteins of similar size; (6) Including competition controls with recombinant RPL26 protein; and (7) Applying appropriate negative controls such as isotype control antibodies (for monoclonals) or pre-immune serum (for polyclonals). Secondary antibody-only controls should always be included to identify non-specific binding from this source .
The observed molecular weight of RPL26 in experimental contexts often differs from its calculated 17 kDa mass, typically appearing between 17-22 kDa on western blots . This variability stems from several factors: (1) Post-translational modifications—ribosomal proteins undergo extensive modifications including phosphorylation, methylation, and ubiquitination that alter electrophoretic mobility; (2) Sample preparation conditions—heat denaturation time and temperature can affect protein migration, with boiling sometimes causing anomalous migration of ribosomal proteins; (3) Gel percentage and buffer systems—higher percentage gels (15-18%) typically provide more accurate sizing for small proteins like RPL26; (4) Protein-protein interactions that resist denaturation; (5) Protein isoforms generated through alternative splicing; and (6) Species-specific sequence variations affecting migration. Researchers should validate observed band patterns using positive controls with known RPL26 expression and consider complementary approaches such as mass spectrometry for definitive identification when results deviate from expected patterns .
When multiple bands appear in Western blots probed with RPL26 antibodies, systematic analysis is required for accurate interpretation: (1) Compare observed bands with predicted molecular weight (17 kDa) and documented range (17-22 kDa) ; (2) Evaluate consistency across different cell lines—true RPL26 signal should maintain similar patterns across validated cell types like K-562, HeLa, HepG2, and Jurkat cells ; (3) Perform verification with multiple antibodies targeting different RPL26 epitopes; (4) Conduct siRNA or CRISPR knockdown experiments to confirm which bands diminish with reduced RPL26 expression; (5) Consider post-translational modifications that may generate multiple forms of the protein; (6) Analyze subcellular fractions separately (nuclear, cytoplasmic, membrane) to determine compartment-specific variants; (7) Compare reducing versus non-reducing conditions to identify potential disulfide-linked complexes; and (8) Evaluate proteasome inhibitor treatment effects to identify potential degradation products. Additional bands at higher molecular weights might indicate ubiquitinated forms, while lower bands could represent proteolytic fragments .
RPL26 antibodies provide valuable tools for analyzing ribosome composition and dynamics in translational research. Advanced applications include: (1) Polysome profiling with subsequent immunoblotting to track RPL26 distribution across monosomes, polysomes, and ribosomal subunits during various cellular conditions; (2) Ribosome immunoprecipitation (IP) to isolate intact ribosomes and analyze associated mRNAs or proteins; (3) Proximity labeling approaches where RPL26 antibodies help validate ribosome-proximity labeling in techniques like BioID or APEX; (4) Sucrose gradient fractionation followed by RPL26 immunoblotting to assess ribosome assembly states; (5) Combining with puromycin labeling to correlate RPL26-containing ribosomes with active translation sites; and (6) Structural studies where antibodies help validate ribosome purification before cryo-EM analysis. These approaches have contributed to understanding translational regulation during stress responses, development, and disease states, with published methodologies available for reference .
Co-immunoprecipitation (Co-IP) with RPL26 antibodies requires special considerations to maintain native interactions and minimize artifacts: (1) Lysis buffer composition is critical—gentler non-ionic detergents (0.5% NP-40 or 1% Triton X-100) help preserve ribosomal integrity compared to stronger ionic detergents; (2) Salt concentration must be carefully optimized—typically 150mM NaCl provides a balance between specificity and maintaining interactions; (3) RNase treatment controls help distinguish RNA-dependent versus direct protein-protein interactions; (4) Antibody orientation strategies—using pre-bound magnetic beads rather than protein A/G sepharose can reduce background; (5) Pre-clearing lysates with beads alone reduces non-specific binding; (6) Washing stringency requires careful optimization to remove non-specific binders while preserving genuine interactions; (7) Elution conditions affect recovery—either gentle (competitive peptide elution) or denaturing (SDS buffer); and (8) Validating results with reciprocal IPs when possible. RPL26 antibodies with demonstrated success in Co-IP applications have been reported in at least one publication, confirming feasibility of this approach .
RPL26 antibodies enable investigation of specialized translational control mechanisms through several sophisticated approaches: (1) Dual immunofluorescence combining RPL26 with translation initiation factors to visualize translation compartmentalization during stress or development; (2) Chromatin immunoprecipitation to study potential extra-ribosomal roles of RPL26 in transcriptional regulation; (3) Ribosome profiling combined with RPL26 immunoprecipitation to identify specifically translated mRNA subsets; (4) Proximity ligation assays (PLA) to detect in situ interactions between RPL26 and regulatory proteins like p53; (5) FRAP (Fluorescence Recovery After Photobleaching) analysis using fluorescently-tagged antibody fragments to study ribosome mobility; (6) Correlative light-electron microscopy using immunogold-labeled RPL26 antibodies to visualize ribosome ultrastructural organization; and (7) Sequential immunoprecipitation approaches to isolate specialized ribosome subpopulations. These methods have revealed RPL26's roles beyond structural ribosome function, including direct interactions with specific mRNAs and regulatory factors during cellular stress responses .
Ribosomal proteins including RPL26 play crucial roles during cellular stress, and specialized antibody applications help elucidate these mechanisms: (1) Pulse-chase immunoprecipitation to track RPL26 synthesis, incorporation, and turnover rates during stress conditions; (2) Nucleo-cytoplasmic fractionation with RPL26 immunoblotting to monitor stress-induced changes in ribosomal protein localization; (3) Immunofluorescence microscopy tracking RPL26 relocalization during nucleolar stress response; (4) Phospho-specific RPL26 antibodies to detect stress-induced post-translational modifications; (5) Co-immunoprecipitation combined with mass spectrometry to identify stress-specific RPL26 interaction partners; (6) Analysis of RPL26 association with stress granules or processing bodies during translational reprogramming; and (7) IRES-dependent translation studies where RPL26 may participate in selective mRNA translation during global translation inhibition. These approaches have revealed emerging non-canonical functions of RPL26 in cellular adaptation to environmental challenges, DNA damage responses, and translation regulation of specific transcripts during stress .
Maintaining RPL26 antibody integrity requires appropriate storage practices to preserve recognition capacity and prevent degradation. Optimal conditions include: (1) Storage temperature at -20°C for long-term preservation, with antibodies remaining stable for approximately one year after shipment under these conditions ; (2) Storage buffer composition typically containing PBS with preservatives such as 0.02% sodium azide and stabilizers like 50% glycerol at pH 7.3 ; (3) Aliquoting considerations—while unnecessary for standard -20°C storage according to some manufacturers, aliquoting prevents repeated freeze-thaw cycles that can compromise antibody performance; (4) Small-volume preparations (20μl) often contain 0.1% BSA as an additional stabilizer ; (5) Avoiding contamination through proper handling and sterile technique; (6) Protection from light for fluorophore-conjugated antibody variants; and (7) Proper labeling including receipt date, dilution information, and freeze-thaw counts to track usage. Following manufacturer-specific recommendations remains crucial, as formulations may vary between suppliers .
Proper handling of RPL26 antibodies before and during experiments significantly impacts their performance: (1) Thawing protocol—gently thaw frozen antibodies at 4°C or room temperature rather than using heat, which can denature antibody proteins; (2) Mixing technique—avoid vortexing which can cause protein denaturation, instead use gentle inversion or pipetting; (3) Centrifugation recommendations—briefly centrifuge thawed antibody vials to collect liquid at the bottom before opening; (4) Dilution practices—prepare working dilutions immediately before use in appropriate buffers (often PBS with 0.1% BSA for blocking); (5) Primary antibody dilutions—WB applications typically use 1:500-1:1000, while IF/ICC applications require 1:50-1:500 dilutions ; (6) Temperature considerations—while room temperature incubations may work, longer incubations at 4°C often improve signal-to-noise ratios; (7) Reuse limitations—while some researchers reuse primary antibody solutions, this practice increases contamination risk and reduces sensitivity with each use; and (8) Optimization requirements—each experimental system may require titration to determine optimal conditions .
Implementing systematic quality control procedures ensures reliable and reproducible results when working with RPL26 antibodies: (1) Positive controls—include cell lines with validated RPL26 expression such as K-562, HeLa, HepG2, or Jurkat cells ; (2) Negative controls—where possible, include RPL26 knockdown/knockout samples or tissues known to lack expression; (3) Experimental replicates—biological and technical replication validates observation consistency; (4) Antibody validation records—maintain documentation of validation experiments including Western blots showing expected 17-22 kDa band pattern ; (5) Lot-to-lot consistency testing when receiving new antibody batches; (6) Cross-validation using alternative detection methods or antibodies targeting different epitopes; (7) Reference standard inclusion—where possible, include recombinant RPL26 protein as a molecular weight reference; and (8) Regular assessment of antibody performance over time to detect potential degradation. These approaches collectively minimize experimental variability and enhance data reliability, particularly important for quantitative applications or when comparing results across different experimental conditions .