RPL22 Human

Ribosomal Protein L22 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Production

RPL22 Human is a 151-amino-acid polypeptide (1–128 residues) with a molecular mass of 17.0 kDa, excluding a 23-amino-acid N-terminal His-tag introduced in recombinant production . Key features include:

PropertyDetail
Gene LocationChromosome 1p36.3-p36.2
ExpressionUbiquitous in human tissues, with nucleolar localization
Recombinant ProductionExpressed in E. coli; purified via chromatography; stored at -20°C

The initiating methionine is post-translationally removed, and the protein binds Epstein-Barr virus (EBV)-encoded RNAs (EBER1/2) and heparin .

Functional Roles Beyond Ribosome Biogenesis

While traditionally associated with translation, RPL22 exhibits non-canonical functions:

RNA Binding and Splicing Regulation

RPL22 interacts with viral RNAs (e.g., EBERs) and modulates RNA splicing. Studies in zebrafish and mice show its role in altering exon usage patterns . For example:

  • Mechanism: Binds intronic hairpin structures to repress splicing, as seen in RPL22B autoregulation .

  • Pathway Impact: Alters proteins involved in RNA processing (e.g., KAP1, HP1γ) and heterochromatin structure .

Disease-Associated Functions

RPL22’s dysregulation is implicated in multiple pathologies:

DiseaseMechanismSource
T-ALLInactivation linked to reduced survival; accelerates leukemia in MyrAkt2 mice
Stem Cell AgingOverexpression disrupts nucleolar heterochromatin, increasing rRNA expression
PsoriasisPromotes keratinocyte proliferation via CyclinD1 and CD4+ T-cell chemotaxis
Insecticide ResistanceOverexpression in deltamethrin-resistant Culex mosquitoes (paradoxical effects)

Stem Cell Aging

A 2024 CRISPR/Cas9 screen identified RPL22 as a key driver of human mesenchymal progenitor cell aging. Key findings:

  • Pro-aging Effect: Overexpression accelerates aging phenotypes (e.g., UV/H₂O₂-induced stress) .

  • Mechanism: Nucleolar localization disrupts heterochromatin (reduces KAP1, HP1γ, H3K9me3), increasing rRNA synthesis .

  • Therapeutic Potential: RPL22 knockout alleviates aging in models of progeria and physiological aging .

T-ALL and Ribosome Heterogeneity

  • RPL22 Deficiency: Impairs T-lymphocyte development and enhances leukemia transformation .

  • Paralog Compensation: Rpl22l1 upregulation compensates for Rpl22 loss in mice, maintaining ribosome function .

Psoriasis Pathogenesis

  • Upregulated Expression: Observed in psoriatic skin lesions; histone H3K27 hyperacetylation drives transcription .

  • Pathological Roles:

    • Proliferation: Increases CyclinD1, promoting keratinocyte S-phase entry .

    • Inflammation: Induces CXCL10 expression, recruiting CD4+ T cells .

Table 1: Molecular Properties of RPL22 Human

ParameterValue
Amino Acids151 (1–128 residues)
Molecular Mass17.0 kDa
Gene ID6146 (Homo sapiens)
SynonymsEAP, HBP15, L22

Table 2: Disease Associations

DiseaseRPL22 RoleModel
T-ALLInactivation accelerates leukemia progressionMyrAkt2 transgenic
PsoriasisDrives keratinocyte proliferation and T-cell recruitmentIMQ-induced mice
Stem Cell AgingDisrupts nucleolar heterochromatinHuman mesenchymal progenitors

Product Specs

Introduction
RPL22, a cytoplasmic ribosomal protein belonging to the L22E family, is found within the 60S ribosomal subunit. This protein exhibits specific binding affinity for Epstein-Barr virus-encoded RNAs (EBERs) 1 and 2.
Description
Recombinant human RPL22, expressed in E.coli, is a single, non-glycosylated polypeptide chain consisting of 151 amino acids (residues 1-128). It has a molecular weight of 17.0 kDa. The protein is engineered with a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The provided RPL22 solution (0.25 mg/ml) is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 1 mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 85% as determined by SDS-PAGE analysis.
Synonyms

Ribosomal Protein L22, Epstein-Barr-Encoded RNA-Associated Protein, Epstein-Barr Virus Small RNA-Associated Protein, 60S Ribosomal Protein L22, EBER-Associated Protein, EAP, HBP15/L22.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAPVKKL VVKGGKKKKQ VLKFTLDCTH PVEDGIMDAA NFEQFLQERI KVNGKAGNLG GGVVTIERSK SKITVTSEVP FSKRYLKYLT KKYLKKNNLR DWLRVVANSK ESYELRYFQI NQDEEEEEDE D.

Q&A

What is RPL22 and what is its primary function in human cells?

RPL22 is a component of the 60S ribosomal subunit that contributes to ribosome structure and function. While ribosomal proteins (RPs) generally don't directly catalyze peptidyl transfer, they play critical regulatory and structural roles in the ribosome . Beyond its canonical ribosomal function, RPL22 has several extra-ribosomal functions, including binding to cellular and viral RNAs outside the ribosomal context . Recent research has identified RPL22 as a heterochromatin destabilizer that promotes cellular senescence .

Experimental approach: RPL22 function can be studied through:

  • Ribosome profiling to assess translational activity

  • Co-immunoprecipitation followed by mass spectrometry to identify interacting partners

  • CRISPR/Cas9-mediated knockout to determine phenotypic consequences

What is the relationship between RPL22 and its paralog RPL22L1?

RPL22 and RPL22L1 (RPL22-like1) are paralogs with highly homologous protein sequences. RPL22 actively represses RPL22L1 expression by binding to a hairpin structure in RPL22L1 mRNA, thereby destabilizing it . When RPL22 is absent, RPL22L1 expression increases and it can be incorporated into ribosomes, suggesting a compensatory mechanism . This relationship demonstrates a novel mechanism by which ribosome composition is regulated through direct repression of one paralog by another .

Research methodology:

  • RNA structure prediction tools (e.g., M-fold) can identify potential binding motifs

  • RNase protection assays confirm direct binding of RPL22 to RPL22L1 mRNA

  • Transcription inhibition assays help determine mRNA stability effects

How do we experimentally differentiate between RPL22's ribosomal and extra-ribosomal functions?

Distinguishing between ribosomal and extra-ribosomal functions requires strategic experimental design:

ApproachMethodologyOutcome Measure
Subcellular fractionationSeparate nucleolar, nucleoplasmic, and cytoplasmic fractionsLocalization patterns of RPL22
Polysome profilingSucrose gradient centrifugationAssociation with active ribosomes
Mutational analysisGenerate mutants that selectively disrupt specific functionsFunction-specific phenotypes
Protein domain mappingExpress truncated proteinsIdentify domains responsible for distinct functions
RNA-binding assaysCLIP-seq, RIP-seqIdentify RNA targets outside ribosomes

Researchers should implement multiple complementary approaches, as single methodologies may not definitively separate these functions.

What mechanisms mediate RPL22's role in heterochromatin regulation?

RPL22 acts as a heterochromatin destabilizer through several interconnected mechanisms. During cellular senescence, RPL22 accumulates in the nucleolus and binds to rDNA regions . This leads to:

  • Interaction with heterochromatin proteins HP1γ and KAP1, as confirmed by co-immunoprecipitation and mass spectrometry

  • Degradation of HP1γ and KAP1, reducing their availability at rDNA regions

  • Consequent loss of H3K9me3 modification, a key heterochromatin mark

  • Heterochromatin decondensation, particularly at rDNA loci

  • Increased accessibility of rDNA to transcription machinery

  • Elevated rRNA synthesis that contributes to cellular senescence

This regulatory pathway provides a molecular link between ribosomal proteins, chromatin structure, and cellular aging.

How does RPL22 contribute to cellular senescence in human cells?

RPL22 has been identified as a key driver of human stem cell senescence through CRISPR/Cas9-based functional screening . Its contribution to senescence involves:

  • Nucleolar accumulation during aging

  • Binding to rDNA regions and displacing heterochromatin factors

  • Destabilization of HP1γ and KAP1, leading to loss of H3K9me3 marks

  • Increased rRNA transcription, which promotes senescence phenotypes

Importantly, RPL22 depletion counteracts senescence in multiple models:

  • Hutchinson-Gilford progeria syndrome (HGPS) human mesenchymal progenitor cells (hMPCs)

  • Werner syndrome hMPCs

  • H₂O₂ or UV-induced senescent cells

  • Primary MPCs from aged adults

Conversely, RPL22 overexpression accelerates senescence in human vascular endothelial cells , demonstrating its causative role in the senescence process.

What are the optimal experimental approaches for studying RPL22 binding to rDNA?

Investigating RPL22-rDNA interactions requires specialized techniques due to the repetitive nature of rDNA and the nucleolar localization of RPL22:

TechniqueApplicationAdvantagesLimitations
ChIP-seqGenome-wide binding profileComprehensive mappingChallenge with repetitive rDNA sequences
ChIP-qPCRTargeted validationQuantitative analysis of specific regionsLimited to known regions
Nucleolar isolationEnrichment of relevant compartmentReduces backgroundPotential loss of transient interactions
IF-FISHCo-localization in situVisualization in cellular contextLower resolution than ChIP
Proximity ligation assayProtein-DNA interactionsSingle-molecule resolutionRequires optimal antibodies
CUT&RUNAlternative to ChIPHigher signal-to-noiseNew technique with variable protocols

Optimization strategies should include:

  • Crosslinking conditions tailored for nucleolar proteins

  • Sonication parameters adjusted for rDNA chromatin structure

  • Bioinformatic pipelines modified for repetitive sequence analysis

  • Controls with RPL22 knockout cells to confirm specificity

How does RPL22-mediated heterochromatin destabilization affect global gene expression?

The impact of RPL22 on heterochromatin extends beyond rDNA, potentially affecting global gene expression through several mechanisms:

  • Direct effects on rRNA transcription and ribosome biogenesis alter translational capacity

  • Changes in HP1γ and KAP1 availability may affect heterochromatin organization genome-wide

  • Altered H3K9me3 distribution could impact expression of genes normally silenced by heterochromatin

  • Downstream signaling cascades activated by RPL22-mediated events may influence transcription factor activity

Research approaches to investigate these effects include:

  • RNA-seq comparing wild-type and RPL22-deficient cells

  • ATAC-seq to assess changes in chromatin accessibility

  • H3K9me3 ChIP-seq to map heterochromatin alterations

  • Proteomic analysis to identify altered signaling pathways

  • Integrated multi-omics approaches to correlate chromatin, transcriptome and proteome changes

What is the mechanistic basis for RPL22's selective binding to RNA hairpin structures?

RPL22 recognizes specific RNA secondary structures, particularly stem-loop (hairpin) structures with distinctive features:

  • A G-C base pair at the neck of the hairpin followed by a U nucleotide serves as the recognition motif

  • This motif has been identified in RPL22L1 mRNA, enabling direct binding and regulation

  • Similar structures are present in viral RNAs like EBER1 from Epstein-Barr virus

Methodological approaches to characterize RNA binding include:

  • RNA structure prediction algorithms (e.g., M-fold)

  • In vitro RNA binding assays with recombinant proteins

  • RNase protection analysis to validate binding to specific structures

  • CLIP-seq to identify binding sites transcriptome-wide

  • Mutational analysis of predicted binding sites to confirm specificity

Understanding this specificity provides insight into how RPL22 selectively regulates target RNAs.

What is the potential of targeting RPL22 to mitigate cellular senescence?

Evidence suggests RPL22 may be a promising target for interventions against cellular senescence:

  • RPL22 depletion counteracts senescence in multiple experimental models:

    • Hutchinson-Gilford progeria syndrome (HGPS) hMPCs

    • Werner syndrome hMPCs

    • H₂O₂ or UV-induced senescent cells

    • Primary MPCs from aged adults

  • Mechanistic rationale: RPL22 inhibition would:

    • Reduce rRNA transcription to normal levels

    • Restore heterochromatin structure at rDNA

    • Normalize HP1γ and KAP1 levels

    • Attenuate senescence-associated phenotypes

Therapeutic targeting strategies could include:

  • RNA interference approaches (siRNA, shRNA)

  • CRISPR/Cas9-based gene editing

  • Small molecule inhibitors of RPL22-DNA or RPL22-RNA interactions

  • Peptide mimetics that disrupt protein-protein interactions

How does RPL22 expression change across different human tissues during aging?

Understanding tissue-specific changes in RPL22 expression is crucial for targeted interventions:

Tissue TypeRPL22 Expression in AgingAssociated Phenotypes
Mesenchymal progenitor cellsIncreasedReduced proliferation, elevated SA-β-Gal activity
Vascular endothelial cellsIncreased upon overexpressionDiminished clonal expansion, higher SA-β-Gal positivity
Multiple organsUpregulated (based on Aging Atlas database)Various age-related phenotypes

Research methodologies to investigate tissue-specific patterns:

  • Single-cell RNA-seq from young vs. aged tissue samples

  • Tissue microarrays with RPL22-specific antibodies

  • Transgenic reporter mice to track RPL22 expression in vivo

  • Comparative proteomics across tissues during aging

The Aging Atlas database indicates RPL22 upregulation in multiple organs during aging, suggesting a conserved role in age-related processes .

What are the optimal CRISPR/Cas9 strategies for RPL22 functional studies?

CRISPR/Cas9 approaches for RPL22 manipulation require careful design:

StrategyApplicationConsiderations
Complete knockoutLoss-of-function studiesMay trigger RPL22L1 compensation
Conditional knockoutTemporal controlRequires Cre-loxP or similar systems
Knockin mutationsDomain-specific functionsPreserves expression levels
CRISPRiTranscriptional repressionReversible, titratable reduction
CRISPRaOverexpression studiesModels pathological increase
Base editingSpecific amino acid changesMinimal DNA damage response

Technical recommendations:

  • Design multiple gRNAs targeting different exons

  • Include controls for off-target effects

  • Verify knockout by sequencing and Western blotting

  • Consider concurrent RPL22L1 manipulation to address compensation

  • Employ rescue experiments with wildtype or mutant RPL22 to confirm specificity

What are the key considerations for identifying RPL22-interacting proteins?

Protein interaction studies for RPL22 should address its dual localization (ribosomal and nucleolar) and multiple functions:

  • Affinity purification approaches:

    • Flag-tagged RPL22 expression followed by co-immunoprecipitation and mass spectrometry

    • Endogenous immunoprecipitation for physiological interactions

    • BioID or APEX2 proximity labeling for transient or compartment-specific interactions

  • Validation strategies:

    • Reciprocal co-immunoprecipitation

    • Domain mapping to identify interaction regions

    • In vitro binding assays with recombinant proteins

    • Functional studies of identified interactions (e.g., shRNA knockdown of interaction partners)

  • Compartment-specific considerations:

    • Subcellular fractionation before interaction studies

    • Nucleolar isolation protocols for nucleolar interactions

    • Polysome purification for ribosome-associated interactions

This comprehensive approach has identified key interactors including heterochromatin proteins HP1γ and KAP1 .

What are the implications of RPL22 research for understanding ribosome heterogeneity?

RPL22's relationship with RPL22L1 provides insight into ribosome heterogeneity and specialized ribosomes:

  • RPL22 directly represses RPL22L1 expression, creating a regulatory mechanism for ribosome composition

  • In RPL22's absence, RPL22L1 is incorporated into ribosomes, potentially conferring distinct properties

  • This mechanism may represent a broader paradigm for how ribosome specificity is coordinated

Future research questions include:

  • Do RPL22-containing and RPL22L1-containing ribosomes preferentially translate different mRNAs?

  • What structural differences exist between these ribosome populations?

  • Are there tissue-specific preferences for RPL22 versus RPL22L1 incorporation?

  • How does ribosome heterogeneity contribute to development, aging, and disease?

How does RPL22 integrate with other aging-related pathways?

RPL22's role in senescence likely intersects with established aging pathways:

  • Potential connections to investigate:

    • DNA damage response pathways, as RPL22 depletion reduces DNA damage signals

    • Nutrient sensing pathways (mTOR, AMPK) that regulate ribosome biogenesis

    • Sirtuin-mediated epigenetic regulation that affects heterochromatin

    • Mitochondrial dysfunction and metabolic reprogramming during senescence

  • Integrated research approaches:

    • Epistasis studies combining RPL22 manipulation with modulation of other pathways

    • Multi-omics approaches to map network interactions

    • Systems biology modeling of aging regulatory networks

    • Comparative studies across model organisms with different aging rates

Understanding these interactions could reveal synergistic targets for intervention in age-related diseases.

Product Science Overview

Introduction

Ribosomal Protein L22 (RPL22) is a component of the large 60S subunit of the ribosome, which plays a crucial role in protein synthesis. Ribosomes are essential macromolecular machines within the cell, responsible for translating mRNA into functional proteins. RPL22 is one of the many ribosomal proteins that contribute to the structure and function of the ribosome.

Structure and Function

RPL22 is an external protein on the 60S ribosomal subunit that is incorporated into the ribosome at later stages of ribosome maturation . It has a highly conserved structure across different species, indicating its essential role in ribosome function. The protein is involved in the assembly and stability of the ribosome, ensuring accurate translation of genetic information.

Regulatory Role

Recent studies have suggested that ribosomal proteins, including RPL22, may have regulatory functions beyond their structural roles. For instance, RPL22 has been shown to control the composition of the ribosome by directly repressing the expression of its own paralog, RPL22-like1 (RPL22L1) . This regulation is achieved through binding to an internal hairpin structure in the mRNA of RPL22L1, thereby influencing its expression and incorporation into ribosomes.

Human Recombinant RPL22

Human recombinant RPL22 is produced using recombinant DNA technology, which involves inserting the gene encoding RPL22 into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the protein for research and therapeutic purposes. Recombinant RPL22 retains the same structure and function as the naturally occurring protein, making it a valuable tool for studying ribosome biology and potential therapeutic applications.

Clinical Significance

Mutations in ribosomal proteins, including RPL22, have been linked to various human diseases. These include developmental malformations, inherited bone marrow failure syndromes, and cancer . Understanding the role of RPL22 in ribosome function and its regulatory mechanisms can provide insights into the molecular basis of these diseases and potential therapeutic targets.

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.