RPL11 Human

Ribosomal Protein L11 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Role in Ribosome Biogenesis

RPL11 is indispensable for LSU formation and rRNA processing. Its dysfunction disrupts ribosomal assembly, leading to:

  • Nucleolar stress: Accumulation of free RPL11 in the nucleoplasm inhibits MDM2, stabilizing p53 and triggering apoptosis .

  • 5S RNP complex formation: Partners with RPL5 and 5S rRNA to mediate p53 activation during ribosomal stress .

Regulation of p53 and c-Myc

RPL11 modulates tumor suppressors and oncogenes:

  • p53 pathway: Binds MDM2, preventing p53 degradation and promoting apoptosis .

  • c-Myc suppression: Inhibits c-Myc transcriptional activity and mRNA stability .

Diamond-Blackfan Anemia (DBA)

Mutations in RPL11 account for ~5% of DBA cases (DBA7). Key characteristics:

FeatureDescription
Mutation TypeMissense, nonsense, frameshift
MechanismRibosomal insufficiency → apoptosis in erythroid precursors → anemia
Associated PhenotypesCraniofacial abnormalities, skeletal defects, cancer predisposition

Table 1: RPL11 Mutations in DBA

MutationEffectReferences
p.Arg98TrpImpaired ribosome function
p.Arg142TrpReduced 5S rRNA binding
p.Ala64ThrDisrupted LSU assembly

Cancer and Chemotherapy Response

RPL11 expression predicts 5-fluorouracil (5-FU) sensitivity in gastric cancer:

  • High RPL11 expression: Better survival in 5-FU-treated patients (P = 0.041) .

  • Mechanism: Enhances p53-dependent apoptosis via MDM2 inhibition .

Table 2: RPL11 and 5-FU Sensitivity in Gastric Cancer

Interaction Network

RPL11 interacts with key regulators of cell cycle and apoptosis:

Partner ProteinFunctionMechanismReferences
MDM2p53 ubiquitinationCompetitive binding to Mdm2
p53Tumor suppressionStabilization via MDM2 inhibition
c-MycOncogenesismRNA degradation, transcriptional inhibition
PMLNucleolar organizationPromotes PML localization

Therapeutic Implications

  • Biomarker potential: RPL11 expression levels may stratify patients for 5-FU therapy .

  • Targeted strategies: Modulating RPL11 to enhance p53 activation in TP53-wildtype cancers .

  • Cancer risk: RPL11 mutations/deletions are observed in breast, melanoma, and multiple myeloma .

Product Specs

Introduction
Ribosomes, the protein synthesis machinery within cells, consist of a small 40S subunit and a larger 60S subunit. Ribosomal Protein L11 (RPL11), a member of the L5P family, is an integral part of the 60S subunit. This protein, located within the cytoplasm, is believed to interact with the 5S rRNA molecule.
Description
Recombinant human RPL11, expressed in E.coli, is a single, non-glycosylated polypeptide chain. It comprises 201 amino acids, including a 23 amino acid His-tag at the N-terminus, and has a molecular weight of 22.6kDa. The purification process involves proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized through filtration.
Formulation
The RPL11 solution is provided at a concentration of 0.5mg/ml. It is buffered with 20mM Tris-HCl (pH 8.0) and contains 0.2M NaCl, 1mM DTT, 2mM EDTA, 250mM Imidazole, and 50% glycerol.
Stability
For short-term storage (up to 4 weeks), the solution should be kept refrigerated at 4°C. For longer periods, it is recommended to store the protein frozen at -20°C. The addition of a carrier protein like HSA or BSA (0.1%) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Analysis by SDS-PAGE confirms a purity level exceeding 90%.
Synonyms
Ribosomal Protein L11, Cell Growth-Inhibiting Protein 34, CLL-Associated Antigen KW-12, 60S Ribosomal Protein L11, DBA7, GIG34, L11.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAQDQGE KENPMRELRI RKLCLNICVG ESGDRLTRAA KVLEQLTGQT PVFSKARYTV RSFGIRRNEK IAVHCTVRGA KAEEILEKGL KVREYELRKN NFSDTGNFGF GIQEHIDLGI KYDPSIGIYG LDFYVVLGRP GFSIADKKRR TGCIGAKHRI SKEEAMRWFQ QKYDGIILPG K

Q&A

What is the structural basis for RPL11's interaction with MDM2?

The crystal structure of human MDM2-RPL11 complex at 2.4 Å resolution reveals that MDM2 extensively interacts with RPL11 through an acidic domain and two zinc fingers. This interaction induces substantial conformational changes in both proteins. The MDM2 protein mimics 28S rRNA binding to RPL11, and the C4 zinc finger specifically determines RPL11 binding to MDM2 but not its homolog MDMX . For structural studies, researchers should consider X-ray crystallography or cryo-EM approaches, preparing highly purified recombinant proteins expressed in bacterial or insect cell systems.

What experimental techniques are most effective for measuring RPL11 expression?

Quantitative real-time PCR is the gold standard for measuring RPL11 mRNA expression. When implementing this method, researchers should:

  • Isolate total RNA using TRIzol reagent following manufacturer protocols

  • Perform reverse transcription (1 μg RNA recommended) using appropriate kits (e.g., ReverTra Ace)

  • Use specific primers for human RPL11 (forward: 5′-GAAAAGGAGAACCCCATGC-3′, reverse: 5′-CATTTCTCCGGATGCCAA-3′)

  • Normalize expression using housekeeping genes such as GAPDH

  • Quantify expression using the comparative cycle threshold method
    Western blotting provides protein-level confirmation, while immunohistochemistry offers spatial context in tissue samples.

How does RPL11 expression influence chemotherapeutic efficacy in gastric cancer?

RPL11 expression is a crucial factor affecting the sensitivity of gastric cancer to 5-fluorouracil (5-FU). High RPL11 expression correlates with better prognosis in 5-FU-treated gastric cancer patients. Mechanistically, RPL11 activates the p53 pathway, including downstream targets P21 and Fas, resulting in suppression of tumor progression. When RPL11 is knocked down in TP53 wild-type gastric cancer cell lines, there is a reversal of the decreased cell viability typically observed with 5-FU treatment . This suggests that:

  • RPL11 expression levels could serve as a biomarker for predicting 5-FU sensitivity in gastric cancer

  • Therapeutic approaches aimed at elevating RPL11 expression might improve 5-FU efficacy in resistant cases

  • RPL11-mediated sensitivity is p53-dependent, as demonstrated in knockdown experiments

What methodological approaches are recommended for investigating RPL11's role in cancer cell viability?

When investigating RPL11's impact on cancer cell viability:

  • Cell viability assays: MTT assay is an established method, exposing cells to varying concentrations of chemotherapeutics (e.g., 5-FU) for 72 hours after RPL11 knockdown or overexpression

  • Gene silencing: Use siRNA transfection targeting different regions of RPL11 mRNA (multiple siRNAs recommended to validate specificity)

  • Pathway analysis: Combine RPL11 manipulation with p53 pathway component knockdowns to establish mechanistic relationships

  • Prognostic assessment: Utilize the Kaplan-Meier plotter online database incorporating multiple gastric cancer datasets (GSE14210, GSE15459, GSE22377, GSE29272, GSE51105, GSE62254) for survival analysis based on RPL11 expression
    Statistical analysis should employ one-way ANOVA followed by post-hoc tests (e.g., Dunnett's test) for multiple group comparisons, with P<0.05 considered statistically significant.

What is the molecular mechanism by which RPL11 activates p53?

The RPL11-mediated p53 activation follows this established mechanism:

  • Under ribosomal stress conditions, RPL11 is released from ribosomes

  • Free RPL11 binds to the central acidic domain and two zinc fingers of MDM2

  • This binding inhibits MDM2's E3 ubiquitin ligase activity toward p53

  • Reduced ubiquitination of p53 leads to its stabilization and accumulation

  • Stabilized p53 activates transcription of target genes involved in cell cycle arrest and apoptosis
    The crystal structure at 2.4 Å resolution demonstrates that formation of the MDM2-RPL11 complex induces substantial conformational changes in both proteins. Importantly, the C4 zinc finger determines RPL11 binding to MDM2 but not to MDMX, providing specificity to this interaction .

How can researchers experimentally validate the RPL11-MDM2-p53 pathway in their cellular models?

Experimental validation of this pathway should include:

  • Co-immunoprecipitation assays to confirm RPL11-MDM2 interaction

  • Proximity ligation assays to visualize the interaction in situ

  • Ubiquitination assays to demonstrate the effect of RPL11 on p53 ubiquitination

  • Reporter assays using p53-responsive promoters (e.g., p21, PUMA)

  • Site-directed mutagenesis of key residues in the interaction interfaces

  • Combined knockdown experiments (RPL11+p53) to confirm pathway dependency

  • Ribosomal stress inducers (actinomycin D at low doses, 5-FU) to activate the pathway
    When interpreting results, researchers should consider that different cell types may exhibit varying dependencies on this pathway based on their p53 status and expression levels of pathway components.

What mechanisms underlie RPL11-associated anemia development?

RPL11 haploinsufficiency leads to anemia through several interconnected mechanisms:

  • Activation of p53 in hematopoietic tissues

  • Impeded erythroid precursor differentiation

  • Insufficient red blood cell development

  • Disrupted ribosomal biogenesis affecting erythropoiesis
    Animal models demonstrate that deletion of one Rpl11 allele in adult mice leads to acute anemia, whereas deletion in germ cells results in embryonic lethality. Mechanistically, reducing p53 dosage through deletion of one p53 allele can mitigate these effects, confirming the p53-dependent nature of RPL11-associated anemia . Additionally, altered coordination between mitochondrial components and RPL11 expression may contribute to the variable expressivity of anemia in affected individuals .

What experimental models are available for studying RPL11 in erythropoiesis?

Researchers studying RPL11's role in erythropoiesis have several models at their disposal:

  • Conditional RPL11-deletion mice allow for tissue-specific and temporal control of RPL11 expression

  • Human CD34+ hematopoietic stem cell cultures can be manipulated using shRNA or CRISPR-Cas9 to reduce RPL11 levels

  • Zebrafish models offer advantages for high-throughput screening and visualization of erythropoiesis

  • Patient-derived cells from individuals with RPL11 mutations provide clinically relevant models
    For robust analysis, researchers should employ:

  • Flow cytometry to assess erythroid differentiation (CD71/CD235a expression)

  • Colony-forming assays to evaluate erythroid progenitor function

  • RNA-seq to identify dysregulated pathways

  • p53 activity assays to confirm pathway involvement

  • Rescue experiments with p53 inhibitors to demonstrate causality

How do RPL11 splice variants impact protein function and disease manifestation?

RPL11 splice variants can significantly alter protein function and contribute to disease phenotypes. A notable example is an RPL11 splicing variant with a late stop codon in the 3' untranslated region (3'UTR) observed in individuals with bilateral triphalangeal thumbs, heart defects, short stature, and anemia .
Research indicates that:

  • RPL11 transcript abundance may be comparable among carriers regardless of symptom severity

  • Coordinated expression between mitochondrial components and RPL11 is lost in carriers, potentially contributing to variable expressivity

  • An antisense RNA product is specifically produced from the RPL11 splice variant-containing allele, suggesting complex regulatory mechanisms
    For comprehensive analysis of RPL11 variants, researchers should combine RNA-seq, RT-PCR, and protein expression studies to characterize the functional consequences of alternative splicing.

What bioinformatic approaches are recommended for identifying and characterizing RPL11 variants?

When analyzing RPL11 genetic and transcript variations:

  • RNA-seq analysis should include:

    • Junction read mapping to identify novel splice sites

    • Differential expression analysis between affected and unaffected individuals

    • Assessment of antisense transcription at the RPL11 locus

    • Coordinated expression analysis with mitochondrial and ribosomal components

  • Variant interpretation should consider:

    • Evolutionary conservation of affected residues or regions

    • Structural impact on RPL11-MDM2 interaction or ribosome incorporation

    • Effect on protein stability and expression

    • Potential creation or disruption of regulatory motifs

  • Functional prediction tools should evaluate:

    • Alternative open reading frames

    • Protein domain disruption

    • RNA secondary structure alterations

    • Potential impact on nonsense-mediated decay pathways

What are the optimal approaches for studying RPL11 in ribosomal stress responses?

Investigating RPL11 in ribosomal stress requires:

  • Stress induction methods:

    • Low-dose actinomycin D (5 nM) specifically inhibits RNA polymerase I

    • 5-FU disrupts rRNA processing

    • Nutrient deprivation or mTOR inhibitors affect ribosome biogenesis

  • Biochemical fractionation:

    • Polysome profiling to assess ribosome assembly

    • Nucleolar/nucleoplasmic/cytoplasmic fractionation to track RPL11 localization

  • Interaction dynamics:

    • FRET-based biosensors to monitor RPL11-MDM2 interaction in real-time

    • Proximity labeling (BioID, APEX) to identify stress-specific interactors

    • Quantitative mass spectrometry for temporal interaction profiling

  • Downstream pathway analysis:

    • ChIP-seq for p53 binding sites after ribosomal stress

    • RNA-seq to assess transcriptional responses

    • Proteomics to identify post-translational modifications
      These approaches allow comprehensive characterization of how RPL11 mediates cellular responses to ribosomal stress.

What statistical considerations are critical when analyzing RPL11 expression in patient cohorts?

When analyzing RPL11 expression in clinical studies:

  • Power analysis should account for:

    • Expected effect size based on preliminary data

    • Patient heterogeneity

    • Treatment variability (especially in cancer studies)

    • Multiple testing corrections for genome-wide approaches

  • Survival analysis should:

    • Use Kaplan-Meier methods with log-rank tests for significance

    • Apply multivariate Cox regression to adjust for confounding factors

    • Consider competing risks where appropriate

    • Calculate hazard ratios with 95% confidence intervals

  • Expression analysis requires:

    • Appropriate normalization strategies (GAPDH is commonly used)

    • Non-parametric methods for non-normally distributed data

    • Correction for batch effects

    • Integration of multiple data types (mRNA, protein, clinical outcomes)

  • For treatment response studies:

    • One-way ANOVA followed by appropriate post-hoc tests (e.g., Dunnett's)

    • Dose-response curve fitting for drug sensitivity experiments

    • P<0.05 is typically considered statistically significant Researchers should use established software like GraphPad Prism (version 8 or later) for statistical analysis and data visualization.

Product Science Overview

Gene and Protein Structure

RPL11 is encoded by the RPL11 gene located on chromosome 1p36.11 in humans . The protein belongs to the L5P family of ribosomal proteins and is found in the cytoplasm . It is known to associate with the 5S rRNA, forming part of the ribosomal structure .

Function

Ribosomes are essential organelles that catalyze protein synthesis. They consist of a small 40S subunit and a large 60S subunit. RPL11 is a component of the 60S subunit and is involved in the assembly and function of the ribosome . It plays a role in the translation of mRNA into proteins, a fundamental process for cell growth and function.

Interactions

RPL11 interacts with several other proteins and molecules within the cell. Notably, it has been shown to interact with:

  • MDM2: RPL11 negatively regulates the oncoprotein MDM2, which is involved in the p53-dependent ribosomal-stress checkpoint pathway .
  • NOP53: Another interaction partner, which is involved in ribosome biogenesis .
  • P53: A well-known tumor suppressor protein, indicating RPL11’s role in cellular stress responses .
Clinical Significance

RPL11 has been implicated in various diseases, particularly cancers. For instance, it has been found to promote the proliferation of non-small cell lung cancer (NSCLC) cells by regulating endoplasmic reticulum stress and autophagy . This highlights its potential as a target for cancer therapy.

Alternative Splicing and Pseudogenes

There are alternative splice variants of RPL11 that encode different isoforms, although these have not been fully characterized . Additionally, multiple processed pseudogenes of RPL11 are dispersed throughout the genome .

Research and Applications

Human recombinant RPL11 is used in various research applications to study its function and interactions. It is also valuable in understanding ribosome biogenesis and the molecular mechanisms underlying diseases associated with ribosomal 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 2024 Thebiotek. All Rights Reserved.