RPL22L1 antibody is an immunological reagent specifically designed to detect the ribosomal protein L22-like 1 (RPL22L1), a constituent of the 60S ribosomal subunit. These antibodies serve as essential tools for researchers investigating the expression and function of RPL22L1 in various biological contexts, particularly in cancer research. RPL22L1 itself belongs to the L22E family of ribosomal proteins and represents a paralog of RPL22 . The protein has gained significant attention in recent years due to its emerging role in cancer progression and treatment resistance.
The development of specific antibodies against RPL22L1 has been crucial for advancing our understanding of this protein's biological significance. These antibodies enable researchers to detect and quantify RPL22L1 expression in various tissues and cell types, providing insights into its distribution and potential functions. As research continues to uncover the importance of RPL22L1 in cancer biology, these antibodies have become increasingly valuable for both basic research and potential clinical applications.
RPL22L1 antibodies have demonstrated utility across multiple experimental applications, making them versatile tools for investigating this protein in diverse research contexts. The primary applications include immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA), and Western blotting, each providing different insights into RPL22L1 expression and function.
Immunohistochemistry represents a particularly valuable application, as evidenced by multiple studies that have utilized RPL22L1 antibodies to assess protein expression in tissue samples. In colorectal cancer research, RPL22L1 antibodies have enabled the visualization of protein localization in both nuclear and cytoplasmic compartments . This dual localization pattern has proven significant, as nuclear RPL22L1 staining in particular has been associated with reduced patient survival (p = 0.003), highlighting the prognostic value of this application .
For Western blot applications, RPL22L1 antibodies allow researchers to detect the protein in cell and tissue lysates, confirming expression levels and providing quantitative data. This application has been particularly useful in experimental studies examining the effects of RPL22L1 knockdown or overexpression on cellular phenotypes . ELISA applications, while less frequently reported in the literature, provide another method for quantitative assessment of RPL22L1 levels in biological samples.
It's worth noting that due to the importance of specific and sensitive detection, some researchers have developed custom RPL22L1 antibodies. For instance, in colorectal cancer studies, researchers created a specific anti-human RPL22L1 polyclonal antibody after finding that commercial antibodies did not provide reliable IHC results in patient tumor samples . This highlights both the technical challenges and importance of antibody validation for accurate RPL22L1 detection.
The application of RPL22L1 antibodies in cancer research has revealed this protein's significant potential as a biomarker across multiple malignancies. In colorectal cancer (CRC), immunohistochemical analysis using RPL22L1 antibodies has demonstrated that the protein is highly expressed in various solid tumors, including renal cell carcinoma, breast cancer, lung cancer, and colon sarcoma . Most notably, elevated RPL22L1 staining was significantly associated with colon adenocarcinoma, and patients with the highest level of RPL22L1 staining exhibited reduced survival rates .
In lung adenocarcinoma (LUAD), RPL22L1 antibody-based detection has confirmed upregulation of this protein compared to normal tissue samples. Kaplan-Meier analysis demonstrated that patients with high RPL22L1 expression had poor prognosis, and multivariate analysis confirmed that RPL22L1 was an independent prognostic factor . The diagnostic value of RPL22L1 for LUAD was verified using receiver operating characteristic (ROC) curve analysis, which indicated an area under the curve of 0.833 (95% CI: 0.787-0.879) .
Hepatocellular carcinoma (HCC) research has similarly benefited from RPL22L1 antibodies, with studies revealing that RPL22L1 is markedly elevated in HCC and contributes to adverse patient survival . Functional studies indicated that RPL22L1 overexpression accelerated cell proliferation, migration, invasion, and sorafenib resistance, suggesting its potential as both a prognostic marker and therapeutic target .
Beyond expression analysis, RPL22L1 antibodies have helped researchers uncover mechanistic insights into how this protein contributes to cancer progression and treatment resistance. In colorectal cancer, RPL22L1 has been linked to 5-Fluorouracil resistance through effects on DNA repair proteins MGMT and MLH1 . In hepatocellular carcinoma, RPL22L1 has been shown to activate ERK to induce atypical epithelial-to-mesenchymal transition (EMT) progress . These findings highlight the value of RPL22L1 antibodies in not only identifying biomarker correlations but also elucidating underlying biological mechanisms.
Research utilizing RPL22L1 antibodies has provided significant insights into the molecular mechanisms through which this protein influences cancer biology. In colorectal cancer, immunoblotting and immunohistochemistry with RPL22L1 antibodies have revealed that this protein regulates the expression of DNA repair genes, specifically MGMT and MLH1 . Elevated RPL22L1 expression increases MGMT levels while decreasing MLH1 expression, potentially explaining the association between RPL22L1 and 5-Fluorouracil resistance in colorectal cancer treatment .
The connection between RPL22L1 and treatment resistance appears to be a consistent theme across multiple cancer types. In hepatocellular carcinoma, RPL22L1 overexpression has been shown to enhance sorafenib resistance through ERK activation . These findings suggest that RPL22L1 may serve as a potential marker to guide precision therapy, particularly for utilizing ERK inhibitors to enhance treatment efficacy in patients with high RPL22L1 expression .
In lung adenocarcinoma, RPL22L1 antibody-based studies have uncovered correlations between RPL22L1 expression and genetic alterations in key oncogenic pathways. The top five mutated genes with significant differences between RPL22L1 high and low expression groups included TP53, KRAS, KEAP1, STK11, and EGFR . Furthermore, research has shown that RPL22L1 enhances the growth and metastasis of lung adenocarcinoma cells by suppressing the MDM2/P53 signaling pathway .
Epigenetic regulation of RPL22L1 has also been investigated, with studies finding that DNA methylation levels of RPL22L1 in lung adenocarcinoma were lower than in normal tissues . The negative correlation between RPL22L1 mRNA expression and DNA methylation levels (r = -0.41) suggests epigenetic regulation as a potential mechanism controlling RPL22L1 expression in cancer .
The development of reliable RPL22L1 antibodies represents a critical advancement in studying this protein's role in cancer biology. Commercial antibodies are typically generated using recombinant RPL22L1 protein or synthetic peptides as immunogens. For instance, Proteintech's RPL22L1 antibody utilizes an RPL22L1 fusion protein (Ag10156) as the immunogen, which is then used to immunize rabbits to produce polyclonal antibodies . After collection, these antibodies undergo antigen affinity purification to enhance specificity .
Validation of RPL22L1 antibodies involves multiple approaches to ensure specificity and sensitivity. Western blotting represents a common validation method, demonstrating that the antibody detects a protein of the expected molecular weight (approximately 15 kDa for RPL22L1) . Immunohistochemistry validation confirms appropriate tissue distribution and subcellular localization patterns, with RPL22L1 antibodies typically showing both nuclear and cytoplasmic staining .
Some researchers have found it necessary to develop custom RPL22L1 antibodies for specific applications, particularly for immunohistochemistry on patient tumor samples. In colorectal cancer research, investigators developed an anti-human RPL22L1 polyclonal antibody after commercial options proved unreliable for IHC in patient samples . This custom antibody was rigorously validated using immunoblotting, immunofluorescent staining, and immunohistochemistry to verify specificity before application in clinical studies .
Cross-reactivity testing represents another important aspect of antibody validation, particularly for antibodies designed to detect multiple related proteins. Invitrogen's anti-RPL22/RPL22L1 antibody, for example, is designed to detect both RPL22 and its highly homologous paralog RPL22L1 . Such dual-specificity antibodies can be valuable for comparative studies but require careful validation to understand their binding characteristics.
The expanding role of RPL22L1 in cancer biology suggests numerous future directions for antibody-based research. Current evidence indicates that RPL22L1 not only serves as a prognostic biomarker but also actively contributes to cancer progression and treatment resistance, making it a potential therapeutic target. The development of more diverse RPL22L1 antibody formats, including monoclonal antibodies with enhanced specificity and antibody-drug conjugates for targeted therapy, represents a promising avenue for future research.
Emerging evidence linking RPL22L1 to immune infiltration in lung adenocarcinoma opens another intriguing research direction . RPL22L1 expression has been found to correlate with immune checkpoint genes, tumor mutation burden (TMB), and microsatellite instability (MSI) . This suggests potential applications for RPL22L1 antibodies in immuno-oncology research, possibly helping to identify patients who might benefit from immunotherapy approaches.
The association between RPL22L1 and treatment resistance mechanisms across multiple cancer types indicates a need for further research into combination therapy approaches. For example, the finding that ERK inhibitors could potentiate sorafenib efficiency in RPL22L1-high HCC cells suggests that combining targeted therapies with standard treatments might improve outcomes for patients with elevated RPL22L1 expression . RPL22L1 antibodies will be essential tools for stratifying patients in such studies.
As single-cell analysis techniques continue to advance, RPL22L1 antibodies compatible with flow cytometry and mass cytometry could enable more detailed studies of RPL22L1 expression in heterogeneous cancer cell populations. This could provide insights into which specific cell subpopulations within tumors express RPL22L1 and how this correlates with cellular phenotypes and treatment responses.
Applications : Immunohistochemistry (IHC)
Review: Immunohistochemistry (IHC) assays determine the overexpression of screened genes. (a) Representative IHC pictures show the expression of INHBA, RPL22L1, and CAPZA1 in tissue splices obtained from the same sample.
RPL22L1 is a 14.6-15 kDa protein that functions as a constituent of the 60S ribosomal subunit. Despite its classification as a ribosomal protein, RPL22L1 appears to have extra-ribosomal functions that influence key cellular pathways. Current research has revealed:
RPL22L1 plays crucial roles in hematopoietic stem cell (HSC) emergence through regulation of Smad1 expression
It exhibits antagonistic functions to its paralog RPL22 in development and disease contexts
RPL22L1 overexpression has been associated with enhanced malignant phenotypes in multiple cancer types, including hepatocellular carcinoma and lung adenocarcinoma
It participates in drug resistance mechanisms by activating ERK signaling and modulating DNA repair pathways
Researchers should note that RPL22L1's functions may be tissue-specific and context-dependent, particularly in development versus disease states.
Based on current validation studies, RPL22L1 antibodies have been successfully employed in multiple research applications:
Note that not all commercial antibodies provide reliable IHC results in patient tumor samples. Several studies have needed to develop custom anti-human RPL22L1 antibodies for IHC applications .
RPL22L1 detection requires careful optimization of protein extraction protocols:
For cell lines: Use RIPA buffer supplemented with protease inhibitors, with particular attention to phosphatase inhibitors if studying phosphorylation states
For tissue samples: Flash-freezing followed by mechanical homogenization in cold lysis buffer yields optimal results
For FFPE samples: Extended antigen retrieval is essential, typically requiring:
When extracting RPL22L1 from ribosomal fractions, use buffer conditions that maintain ribosomal integrity while still releasing associated proteins.
Distinguishing between RPL22L1 and RPL22 requires careful experimental design due to their structural similarities but opposing functions:
Antibody selection: Use antibodies raised against unique epitopes that don't cross-react. Validate specificity with knockdown/knockout controls.
Functional analysis: Design experiments that can detect their opposing functions:
RNA binding studies: Both can bind Smad1 RNA, but with opposing effects on expression, so RNA immunoprecipitation combined with expression analysis can differentiate their activities
Phylogenetic approach: Leverage evolutionary conservation analysis, as zebrafish rpl22 and rpl22l1 are closely related to mammalian orthologs and show similar functional divergence
RPL22L1 has emerged as a significant prognostic biomarker in multiple cancer types:
These findings suggest that immunohistochemical analysis of RPL22L1 could serve as a clinically valuable prognostic tool.
RPL22L1 contributes to drug resistance through multiple mechanisms:
In hepatocellular carcinoma:
In colorectal cancer:
In lung adenocarcinoma:
This suggests that RPL22L1 status should be considered when designing treatment strategies, and inhibition of RPL22L1 or its downstream pathways may overcome drug resistance.
RPL22L1 expression correlates with immune infiltration patterns, suggesting potential immunomodulatory functions:
Researchers studying tumor immunology should consider RPL22L1 status when analyzing immune infiltration patterns and immunotherapy responsiveness.
A robust validation strategy for RPL22L1 antibodies should include:
Positive and negative controls:
Multi-method validation:
Cross-species reactivity:
Application-specific validation:
For advanced applications like ChIP-seq or single-cell protein analysis, additional validation steps specific to those methods should be implemented.
Single-cell analysis of RPL22L1 requires specialized approaches:
Single-cell RNA sequencing (scRNA-seq):
The Tumor Immune Single-cell Hub 2 (TISCH2) database (http://tisch.comp-genomics.org/) provides resources for examining RPL22L1 expression at the single-cell level
Analysis should account for technical variability and dropout events common in scRNA-seq data
Normalization and scaling procedures specific to ribosomal proteins should be applied
Protein-level detection:
Immunofluorescence with signal amplification may be necessary for detecting low-abundance RPL22L1
Consider multiplexed imaging approaches to simultaneously evaluate RPL22L1 and cell type markers
CyTOF (mass cytometry) protocols with RPL22L1 antibodies require metal conjugation optimization
Analytical approaches:
Clustering algorithms should account for the ubiquitous but variable expression of ribosomal proteins
Dimension reduction techniques like UMAP or t-SNE can help visualize cell populations with distinct RPL22L1 expression patterns
Integration with spatial transcriptomics can provide tissue context for single-cell findings
To assess functional outcomes of RPL22L1 manipulation, researchers should consider:
Genetic manipulation approaches:
Functional assays:
Signaling pathway analysis:
In vivo validation:
Each functional assessment should include appropriate controls, including paralog (RPL22) modulation where relevant.
For investigating RPL22L1's impact on immunity:
Computational approaches:
Experimental validation:
Flow cytometry panels for immune profiling of RPL22L1-high versus low tumors
Co-culture systems with immune cells to assess direct effects
CyTOF for high-dimensional immune phenotyping
Spatial proteomics to map immune cell distribution relative to RPL22L1 expression
Clinical correlation: