PELI2 is a 48.8 kDa recombinant protein produced in Escherichia coli, consisting of 443 amino acids (1–420 a.a.) fused with an N-terminal His-tag . It functions as an E3 ubiquitin ligase, catalyzing the covalent attachment of ubiquitin moieties to substrate proteins, and plays critical roles in Toll-like receptor (TLR), interleukin-1 (IL-1), and STING signaling pathways .
TLR/IL-1 Pathways: Facilitates IRAK1 polyubiquitination, enhancing NF-κB and MAPK activation .
STING Pathway:
NLRP3 Inflammasome: Mediates priming via ubiquitination, contributing to IL-1β maturation and LPS-induced lethality .
Stabilizes transcription factors PU.1 and TCF3 via K63-linked ubiquitination, regulating IL-7R expression and early B-cell progenitor proliferation .
Deficiency in Peli2 knockout mice leads to impaired B-cell differentiation and reduced survival under stress hematopoiesis .
Lupus patients exhibit reduced PELI2 levels, correlating with elevated basal interferon signaling .
PELI2 downregulation during viral infection unleashes interferon production, suggesting therapeutic potential .
PELI2 is an E3 ubiquitin ligase that plays important roles in inflammation and immune system regulation. Structurally, PELI2 possesses a C-terminal RING-like domain responsible for ubiquitin ligase activity and a phospho-threonine-binding forkhead-associated (FHA) domain that facilitates substrate binding . These domains are critical for its function in mediating protein-protein interactions and catalyzing the ubiquitination process. The FHA domain specifically has been demonstrated to interact with the PEST domain of transcription factors like PU.1, enabling PELI2 to regulate their stability .
PELI2 acts as a critical mediator for innate immunity through multiple signaling pathways, including those initiated by IL-1 receptors, Toll-like receptors, and NOD-like receptors . Research indicates that PELI2 preferentially inhibits IRF3 signaling while enhancing NF-κB signaling in the context of the STING pathway . This dual regulatory role appears to be human-specific and represents an evolutionary adaptation that makes the human cGAS-STING pathway more selective in its response to pathogens compared to mice . In early B-cell development, PELI2 regulates IL-7R expression through interactions with transcription factors, which is essential for lymphocyte proliferation and differentiation .
PELI2 is required for early B-cell development, particularly in the transition from common lymphoid progenitors (CLP) to B-cell lineage. Research using conditional knockout mouse models has demonstrated that PELI2 deletion results in:
Significant reduction in B-lineage progenitor cells (BLP, Ly6D+) derived from CLP
Decreased colony formation in IL-7-supplemented culture conditions
Impaired proliferation of early B-cell progenitors without major effects on cell survival
Reduced expression of key B-cell development transcription factors including EBF1, FOXO1, and PAX5
These effects are primarily mediated through PELI2's regulation of IL-7R expression, which is crucial for B-cell proliferation and differentiation .
Researchers investigating PELI2 have successfully employed several experimental models:
Conditional Knockout Mouse Model: PELI2 floxed mice (PELI2 fl/fl) with loxP sites flanking exon 2 crossed with Vav-Cre transgenic mice to generate hematopoietic-specific PELI2 knockout mice .
Inducible Knockout System: PELI2 fl/fl mice crossed with Ubc-cre-ERT2 mice for tamoxifen-inducible deletion of PELI2 in adult mice to study temporal effects .
Competitive Bone Marrow Transplantation: Used to assess PELI2's role in hematopoietic stem cell (HSC) function and self-renewal capacity .
In vitro Cell Culture Systems: Including BCP-ALL cell lines for studying PELI2's role in leukemia .
Xenograft Models: Including NSG mice injected with Nalm-6 cells to study PELI2's role in leukemia progression in vivo .
When designing experiments, researchers should consider both cell-autonomous effects and the influence of PELI2 on the bone marrow microenvironment.
PELI2 regulates protein stability through K63-linked polyubiquitination rather than the K48-linked ubiquitination typically associated with proteasomal degradation. The molecular mechanism includes:
Direct Protein Interaction: PELI2 interacts with target proteins through its FHA domain. For example, the FHA domain of PELI2 binds to the PEST domain of PU.1 .
K63-Polyubiquitination: PELI2 catalyzes K63-linked ubiquitination of target proteins such as PU.1, which enhances protein stability rather than marking proteins for degradation .
Protection from Degradation: PELI2 overexpression protects targets like PU.1 from time-dependent degradation after cycloheximide treatment .
Enhanced Transcriptional Activity: PELI2-mediated stabilization of transcription factors like PU.1 enhances their chromatin occupancy at target gene promoters, including the IL-7R promoter .
This non-degradative ubiquitination represents an important post-translational regulatory mechanism for controlling gene expression programs in immune cell development.
PELI2 functions as a key regulator of the STING pathway in humans by:
Differential Regulation of Downstream Pathways: PELI2 preferentially inhibits IRF3 signaling while enhancing NF-κB signaling downstream of STING activation .
Species-Specific Regulation: PELI2 regulation appears to be specific to human STING but not mouse STING, representing an evolutionary divergence .
Increased Pathway Selectivity: The human-specific PELI2 regulation makes the human cGAS-STING pathway less reactive but more selective to pathogen detection compared to mice .
This species-specific regulation adds to other known differences between human and mouse STING pathways, including differential binding affinity for cyclic dinucleotides and sensitivity to dsDNA stimulation .
PELI2 has been implicated in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) through several mechanisms:
IL-7R Regulation: PELI2 promotes TCF3 protein stability via K63-polyubiquitination to regulate IL-7R expression, which is required for the proliferation of BCP-ALL cells .
Anti-apoptotic Effects: Although the detailed mechanisms require further investigation, PELI2 appears to support BCP-ALL cell survival.
Xenograft Models: Studies using BCP-ALL xenograft models have demonstrated the importance of PELI2 in leukemia progression .
These findings suggest that PELI2 could be a potential therapeutic target in B-cell malignancies, particularly those dependent on IL-7R signaling.
When designing PELI2 knockout experiments, researchers should include:
Multiple Targeting Strategies: Use both conditional (tissue-specific) and inducible knockout systems to distinguish between developmental versus acute effects of PELI2 deletion .
Rescue Experiments: Perform genetic rescue experiments by re-expressing PELI2 or downstream effectors (e.g., PU.1 or IL-7R) to confirm phenotype specificity .
Domain-Specific Mutants: Include PELI2 constructs with mutations in the FHA or RING domains to dissect domain-specific functions .
Time-Course Analysis: Especially important for hematopoietic studies to capture both immediate and long-term consequences of PELI2 deletion .
Competitive Transplantation: Essential for distinguishing cell-intrinsic from microenvironmental effects in hematopoietic studies .
For investigating PELI2's E3 ligase activity, researchers should consider:
Co-Immunoprecipitation (Co-IP): To detect protein-protein interactions between PELI2 and its substrates .
Ubiquitination Assays: Using antibodies specific for K63- versus K48-linked ubiquitin chains to distinguish between degradative and non-degradative ubiquitination .
Cycloheximide Chase Assays: To measure protein stability and half-life in the presence or absence of PELI2 .
Truncation and Domain Mapping: Creating a series of truncated forms of both PELI2 and potential substrates to map interaction domains precisely .
Chromatin Immunoprecipitation (ChIP): To assess the effect of PELI2-mediated stabilization on transcription factor binding to target gene promoters .
E3 Ligase Database Resources: Utilize comprehensive databases of human E3 ubiquitin ligases for comparative studies and identification of potential functional redundancy .
Given the documented differences in PELI2 function between humans and mice, researchers should:
Use Appropriate Models: Consider human cell lines or humanized mouse models when studying PELI2 functions that may not be conserved in mice .
Comparative Analysis: Perform side-by-side comparisons of PELI2 function in human versus mouse cells to identify species-specific mechanisms .
Sequence Analysis: Compare the conservation of key domains and potential regulatory motifs between species .
Pathway Context: Consider that PELI2's effects may differ depending on the cellular context and specific signaling pathway being studied .
Evolutionary Perspective: Frame research questions with awareness that human PELI2 regulation may represent an evolutionary adaptation for more selective immune responses .
Several areas of PELI2 research remain incompletely understood:
Tissue-Specific Functions: While PELI2's role in B cells has been studied, its function in other immune and non-immune cell types requires further investigation.
Substrate Specificity: The complete repertoire of PELI2 substrates beyond PU.1 and TCF3 remains to be identified .
Regulation of PELI2 Activity: The mechanisms controlling PELI2 expression and enzymatic activity in different cellular contexts are not fully elucidated.
Relationship to Other Pellino Family Members: The potential functional redundancy or cooperative relationships between PELI2 and other Pellino family proteins need clarification.
Role in Human Diseases: Beyond BCP-ALL, PELI2's involvement in other inflammatory or immune-related disorders requires investigation.
Future studies of PELI2 could benefit from several cutting-edge approaches:
CRISPR-Based Screening: Genome-wide or targeted CRISPR screens to identify additional PELI2 interactors or regulatory pathways.
Proximity Labeling: BioID or APEX2-based approaches to comprehensively identify the PELI2 interactome in different cellular contexts.
Single-Cell Technologies: scRNA-seq and CyTOF to understand cell-specific effects of PELI2 in heterogeneous populations like bone marrow.
Structural Biology: Cryo-EM or crystallography studies of PELI2 in complex with its substrates to understand the molecular basis of specificity.
Systems Biology: Integration of transcriptomic, proteomic, and ubiquitinomic data to build comprehensive models of PELI2 function in different contexts.
Based on current knowledge, several strategies for therapeutic targeting of PELI2 could be explored:
Small Molecule Inhibitors: Development of compounds that disrupt PELI2's E3 ligase activity or specific protein-protein interactions.
Degrader Technologies: PROTAC or molecular glue approaches to induce PELI2 degradation in specific disease contexts.
Gene Therapy: Approaches to modulate PELI2 expression in conditions where its activity contributes to pathology.
Combination Therapies: Targeting PELI2 in combination with existing therapies for B-cell malignancies, particularly those affecting IL-7R signaling .
Immunomodulation: Leveraging PELI2's role in the STING pathway to enhance or dampen specific aspects of immune responses .
The development of these approaches requires further research into PELI2's tissue-specific functions and potential off-target effects of PELI2 inhibition.
Pellino E3 Ubiquitin Protein Ligase Family Member 2 (PELI2) is a protein encoded by the PELI2 gene in humans. This protein is part of the Pellino family of E3 ubiquitin ligases, which play a crucial role in the regulation of immune responses through the ubiquitylation of specific substrate proteins .
PELI2 is characterized by an N-terminal forkhead-associated (FHA) domain and a C-terminal RING-like domain. The FHA domain recognizes phospho-threonine residues, facilitating the association with interleukin-1 receptor-associated kinases (IRAKs), while the RING-like domain confers E3 ubiquitin ligase activity . This activity involves catalyzing the covalent attachment of ubiquitin moieties onto substrate proteins, a process essential for various cellular functions, including protein degradation, signal transduction, and DNA repair .
PELI2 is implicated in the Toll-like receptor (TLR) and interleukin-1 (IL-1) signaling pathways. It interacts with complexes containing IRAK kinases and TNF receptor-associated factor 6 (TRAF6), mediating the polyubiquitination of IRAK1. This modification is crucial for the activation of downstream signaling pathways, including the mitogen-activated protein kinase (MAPK) pathway and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway . These pathways are vital for the innate immune response, leading to the production of pro-inflammatory cytokines and other immune mediators .
PELI2’s role in immune signaling highlights its importance in both innate and adaptive immunity. By regulating the ubiquitylation of key signaling proteins, PELI2 ensures the proper activation and termination of immune responses. Dysregulation of PELI2 activity has been associated with various immune-related disorders, including hyperlipoproteinemia type IV and 3MC syndrome .
Given its central role in immune signaling, PELI2 is a potential target for therapeutic interventions aimed at modulating immune responses. Research into the molecular mechanisms of PELI2 function and regulation continues to provide insights into its potential as a therapeutic target for treating immune-related diseases .