Recombinant Xenopus laevis Zinc finger protein 36, C3H1 type-like 2-B (zfp36l2-B) is a protein that, in Homo sapiens (humans), is known as ZFP36L2 (ZFP36 ring finger protein like 2) . ZFP36L2 is an RNA-binding protein . It can be purchased as a recombinant protein produced in Yeast, E. coli, Baculovirus, or Mammalian cells .
| Code | Source | Conjugate |
|---|---|---|
| CSB-YP773869XBE | Yeast | N/A |
| CSB-EP773869XBE | E. coli | N/A |
| CSB-EP773869XBE-B | E. coli | Avi-tag Biotinylated E. coli biotin ligase (BirA). Biotin is covalently attached to the 15 amino acid AviTag peptide via an amide linkage, a process catalyzed by BriA through AviTag-BirA technology. |
| CSB-BP773869XBE | Baculovirus | N/A |
| CSB-MP773869XBE | Mammalian cell | N/A |
ZFP36L2 is involved in various biological processes, including mRNA degradation, immune response, and cell cycle regulation .
mRNA destabilization ZFP36L2, an RNA-binding protein, recognizes adenine-uridine-rich elements (ARE) at the 3′UTRs of target mRNAs, leading to mRNA destabilization or translation inhibition .
Immune response ZFP36L2 is associated with T cells and macrophages in the tumor microenvironment . It is highly expressed in CD8+ T cells and IL1β+ macrophages .
Regulation of regulatory T cells ZFP36L2 downregulates helios expression and suppresses the function of regulatory T cells .
Cell cycle regulation ZFP36L2 is a cell cycle-regulated CCCH-domain protein, with its abundance regulated post-transcriptionally .
Association with mechanical stress In Mus musculus (mice), the Zinc finger protein 36 like 2-histone deacetylase 1 axis is involved in bone responses to mechanical stress .
Role in muscle tissue Long noncoding RNA ZFP36L2-AS facilitates intramuscular fat deposition, activates fast-twitch muscle phenotype, and induces muscle atrophy .
Prognostic marker ZFP36L2 may be a potential prognostic marker of IL1β+ osteosarcoma . High expression of ZFP36L2 correlates with the prognosis and immune infiltration in lower-grade glioma (LGG) .
3.1 Impact on Spleen Cells
In a study using L2-fKO mice, the absence of Zfp36l2 did not lead to compensatory increases in other family members like Ttp (Zfp36) and Zfp36l1 . RNA-seq analysis revealed 549 upregulated and 603 downregulated mRNAs in ZFP36L2-lacking cells compared to wild type .
| Category | Number of mRNAs |
|---|---|
| Upregulated | 549 |
| Downregulated | 603 |
| Unchanged | 15,684 |
Three genes (Apol11b, Elavl2, and Fgf23) found to be upregulated by RNA-seq analysis were also significantly increased in RT-qPCR analysis .
3.2. ZFP36L2 Binding Specificity
ZFP36L2-binding requires a minimum of a 7-mer (UAUUUAU), and a probe containing a single 5-mer (AUUUA) seems to be insufficient . There is a statistically significant enrichment of the 7-mer motif in the upregulated genes in the L2-fKO spleen .
4.1. Correlation with PD-L1
ZFP36L2 expression is significantly positively correlated with PD-L1 expression . Patients with low ZFP36L2 expression had significantly prolonged survival in the anti-PD-L1 cohort (IMvigor210) treated with atezolizumab . The proportion of patients with better efficacy for immunotherapy in the ZFP36L2 low-expression group was significantly higher compared to the high-expression group .
Zinc-finger RNA-binding protein that destabilizes several cytoplasmic AU-rich element (ARE)-containing mRNA transcripts. This destabilization occurs through the promotion of poly(A) tail removal (deadenylation), providing a mechanism for attenuating protein synthesis. It acts as a 3'-untranslated region (UTR) ARE mRNA-binding adapter protein, linking signaling events to the mRNA decay machinery. Its function involves recruiting the CCR4-NOT deadenylase complex and potentially other components of the cytoplasmic RNA decay machinery to bound ARE-containing mRNAs, thereby promoting ARE-mediated mRNA deadenylation and decay. It binds to the 3'-UTR ARE of numerous mRNAs and induces ARE-containing mRNA degradation even without a poly(A) tail. It is also required for tubulogenesis during pronephros development.
KEGG: xla:398103
UniGene: Xl.476
ZFP36L2 functions as an RNA-binding protein by:
Recognizing and binding to AU-rich elements (AREs) in the 3'-untranslated regions (UTRs) of target mRNAs through its tandem CCCH zinc finger motifs
Recruiting cellular mRNA decay machinery, primarily through the 5'-3' XRN1-mediated RNA decay pathway
Promoting deadenylation of target transcripts, which leads to their destabilization and degradation
Recent research has demonstrated that unlike ZFP36L1, which recruits both 5'-3' XRN1 and 3'-5' RNA exosome pathways, ZFP36L2 impedes target mRNA stability primarily through the 5'-3' XRN1-mediated decay pathway . This indicates that although these related proteins serve similar functions, they employ distinct mechanisms to regulate RNA stability.
ZFP36L2 expression in Xenopus shows distinct tissue-specific and developmental patterns. In situ hybridization studies at embryonic day 14.5 (E14.5) revealed highest expression in the fetal liver, with detectable expression in many other tissues including the yolk sac . This expression pattern aligns with its critical role in hematopoiesis.
Expression data from Xenopus developmental studies shows:
This expression pattern correlates with ZFP36L2's multiple roles in development, including organogenesis and cell differentiation pathways.
ZFP36L2 serves as a critical regulator of definitive hematopoiesis, with knockout studies demonstrating its essential role in blood cell formation. Complete ZFP36L2 deficiency in mice resulted in:
Early postnatal death (within approximately 2 weeks) due to hemorrhage
Pancytopenia (reduced numbers of red cells, white cells, and platelets)
Significantly reduced definitive multilineage and lineage-committed hematopoietic progenitors
Inability of hematopoietic stem cells to reconstitute the hematopoietic system in irradiated recipients
Quantitative analysis of peripheral blood from ZFP36L2 knockout mice showed:
| Parameter | Wild-type | ZFP36L2 KO | % Reduction |
|---|---|---|---|
| Red blood cells | Normal | Decreased | Significant |
| White blood cells | Normal | Decreased | Significant |
| Hemoglobin | Normal | Decreased | Significant |
| Hematocrit | Normal | Decreased | Significant |
| Platelets | Normal | Decreased | Significant |
These findings establish ZFP36L2 as an essential modulator of hematopoiesis through its regulation of mRNA stability in hematopoietic stem and progenitor cells .
ZFP36L2 plays a crucial role in cell cycle regulation, particularly at the G2-M checkpoint. Recent studies have revealed that:
ZFP36L2 controls progression into mitosis by regulating key cell cycle genes
Deletion of both ZFP36L1 and ZFP36L2 causes accumulation of cells in G2-M phase
ZFP36L2 regulates protein expression of cyclin B1 and CDK1, which together are essential for G2-to-M transition
Loss of ZFP36L2 leads to faster entry into G2 but causes G2-M arrest and increased cell death
Mechanistically, ZFP36L2 binds to mRNAs encoding cell cycle regulators without necessarily changing their abundance but regulates their translation. Flow cytometry analysis of cells with ZFP36L1/ZFP36L2 deletion showed increased protein levels of cyclin B1 and CDK1 despite unchanged mRNA levels, indicating post-transcriptional regulation .
ZFP36L2 has recently been identified as an important component of host antiviral defense, particularly against flaviviruses. Key findings include:
ZFP36L2 restricts Japanese encephalitis virus (JEV) replication by destabilizing viral RNA
Overexpression of ZFP36L2 significantly decreases viral RNA levels as demonstrated by immunofluorescence assay and RT-PCR
ZFP36L2 impairs viral RNA stability through the 5'-3' XRN1-mediated RNA decay pathway
Unlike ZFP36L1, which recruits both 5'-3' XRN1 and 3'-5' RNA exosome pathways, ZFP36L2 primarily utilizes the 5'-3' XRN1 pathway
Experimental evidence showed that cells overexpressing ZFP36L2 exhibited significantly decreased JEV RNA levels compared to control cells expressing EGFP. Western blotting revealed marked reduction in JEV NS3 protein levels in cells overexpressing ZFP36L2 .
Several methodologies have proven effective for analyzing ZFP36L2-RNA interactions:
RNA Immunoprecipitation (RIP): This approach can identify mRNAs bound by ZFP36L2 in cellular contexts. RIP followed by RT-PCR analysis can quantify binding to specific target transcripts .
Individual-nucleotide resolution Cross-Linking and Immunoprecipitation (iCLIP): This advanced technique provides high-resolution mapping of ZFP36L2 binding sites on target mRNAs, identifying precise crosslinking sites with nucleotide-level resolution .
Electrophoretic Mobility Shift Assays (EMSA): These assays demonstrate direct binding between ZFP36L2 and target RNA sequences, particularly useful for analyzing interactions with specific regions like 3'UTRs .
Biotinylated RNA Pull-down: This approach uses in vitro transcribed, biotin-labeled RNA to pull down associated proteins from cell lysates, allowing verification of endogenous ZFP36L2 binding to specific transcripts .
UV Cross-linking Assays: These assays identify direct RNA-protein interactions by UV-mediated crosslinking followed by immunoprecipitation and RNase treatment. The remaining protein-protected RNA fragments can be radiolabeled and analyzed by SDS-PAGE .
For comprehensive analysis, combining multiple approaches is recommended. For example, initial iCLIP to identify binding sites followed by EMSA or biotinylated RNA pull-down to confirm direct interactions .
ZFP36L1 and ZFP36L2 display both redundant and specific functions across various biological contexts:
T cell homeostasis: T cell-specific deletion of any two ZFP36 family genes does not cause spontaneous inflammatory syndrome, while deletion of all three (ZFP36, ZFP36L1, ZFP36L2) results in lethal inflammation
B cell development: ZFP36L1 and ZFP36L2 have redundant roles, and their combined deletion causes severe developmental defects
Target binding: Both proteins bind similar AU-rich elements in target mRNAs