ZRSR2 (zinc finger CCCH-type, RNA-binding motif and serine/arginine-rich protein 2) antibodies are essential tools for studying the role of ZRSR2 in RNA splicing, particularly its involvement in the minor (U12-type) spliceosome. These antibodies enable researchers to detect ZRSR2 protein expression, localization, and interactions in cellular and disease models, especially in myeloid malignancies like myelodysplastic syndromes (MDS) .
ZRSR2 antibodies have been instrumental in demonstrating that ZRSR2 loss disrupts U12-dependent splicing, leading to intron retention in genes like PTEN, BRAF, and MAP kinases. This defect is a hallmark of ZRSR2-mutated MDS . For example:
Knockdown models: ZRSR2-deficient TF-1 and K562 cells show impaired splicing of U12-type introns, validated by RNA-Seq and qRT-PCR .
Patient samples: MDS bone marrow with ZRSR2 mutations exhibits aberrant intron retention, detectable via ZRSR2 antibody-based assays .
Studies using ZRSR2 antibodies revealed that ZRSR2 loss alters myeloid and erythroid differentiation:
In vitro differentiation: ZRSR2 knockdown in CD34+ hematopoietic stem cells reduces erythroid colonies (BFU-E) and increases myeloid (CFU-M) colonies .
Cell cycle effects: ZRSR2-deficient cells show reduced S-phase entry and tumorigenic potential in murine xenograft models .
ZRSR2 antibodies helped identify sex-biased mutation patterns in MDS and blastic plasmacytoid dendritic cell neoplasm (BPDCN):
X-linked tumor suppressor: ZRSR2 mutations occur predominantly in males due to its X-chromosome location and escape from X-inactivation .
Cooperative mutations: ZRSR2 loss synergizes with TET2 mutations to impair dendritic cell activation and cytokine production (e.g., IFNα, IL6) .
Abcepta (AP20436b): Validated in WB using 293, HepG2, and Jurkat cell lysates (35 µg/lane) .
Abcam (ab223062): Confirmed specificity in human K562 and HEK-293 lysates (58 kDa band) and mouse heart tissue .
Novus Biologicals (NBP1-57317): Demonstrated staining in human alveolar and renal tubule epithelial cells .
While ZRSR2 antibodies are critical for spliceosome research, challenges include:
Cross-reactivity: Limited evidence for non-human primate or rat models .
Functional redundancy: Murine Zrsr1 compensates for Zrsr2 loss, complicating in vivo studies .
Future studies should leverage CRISPR-edited models and high-resolution imaging to map ZRSR2 dynamics during spliceosome assembly.
CSB-PA618014LA01HU is a rabbit polyclonal antibody generated against a peptide encompassing amino acids 191-372 of the human ZRSR2 protein. This unconjugated IgG antibody exhibits high specificity for ZRSR2, a crucial splicing factor involved in the recognition of 3' intron splice sites during the early stages of spliceosome assembly. Thoroughly tested for its suitability in ELISA, WB, IHC, and IF applications, this antibody demonstrates a purity exceeding 95% achieved through protein G purification. It demonstrates reactivity with both human and mouse samples.
ZRSR2 is a spliceosomal protein that plays a pivotal role in the splicing of U12-type introns, while having minimal effect on U2-dependent splicing. It contains a CCCH-type zinc finger domain, an RNA-binding motif, and serine/arginine-rich regions that are characteristic of splicing factors . ZRSR2 interacts with several splicing machinery components, including members of the SF3B complex, U2AF1, U2AF2, and SRPK1, forming a functional network essential for proper pre-mRNA processing . Loss of ZRSR2 function results in impaired splicing of U12-type introns, which represent a minor but functionally significant subset of introns in the human genome. This splicing defect can lead to altered gene expression patterns that affect cellular differentiation and proliferation, particularly in hematopoietic cells .
To ensure specific binding when using ZRSR2 antibodies, employ multiple validation approaches:
Use appropriate negative controls, including isotype controls and samples where ZRSR2 is knocked down via shRNA (as demonstrated in studies where ZRSR2 sh1 and sh2 vectors resulted in efficient downregulation) .
Confirm specificity by testing the antibody against recombinant ZRSR2 protein or ZRSR2-overexpressing cells alongside wild-type cells.
Perform competition assays using the immunizing peptide to block specific binding sites.
When conducting Western blotting, verify that the detected band aligns with the expected molecular weight of ZRSR2 (~482 amino acids) .
Cross-validate results using multiple antibodies targeting different epitopes of ZRSR2, such as those directed against the C-terminal region (AA 453-482) versus mid-region epitopes (AA 191-372) .
ZRSR2 antibodies are suitable for various research applications based on their validated reactivity profiles:
Different antibodies show varying cross-reactivity profiles, with some reacting against human ZRSR2 exclusively, while others recognize homologs across multiple species including mouse, rat, horse, dog, cow, rabbit, guinea pig, and monkey .
ZRSR2 mutations in myelodysplastic syndromes (MDS) cause specific splicing aberrations predominantly affecting U12-type introns. These mutations are typically inactivating alterations (nonsense, frame-shift, and splice site mutations) that result in loss of ZRSR2 function .
The splicing defects can be characterized using ZRSR2 antibodies through:
Differential splicing analysis: Compare splicing patterns between ZRSR2 mutant and wild-type samples using RT-PCR and RNA-seq with immunoprecipitation of ZRSR2-associated transcripts. Analysis of MDS bone marrow samples with ZRSR2 mutations revealed 689 mis-spliced junctions, with a striking overabundance of retained U12-type introns .
Splicing complex composition studies: Immunoprecipitation with ZRSR2 antibodies coupled with mass spectrometry identified key ZRSR2 interacting partners including SF3B complex members, U2AF1, U2AF2, and SRPK1, as well as novel partners such as C1QBP and CCDC97 .
Functional validation: ZRSR2 antibodies can validate knockdown efficiency in experimental models designed to recapitulate MDS splicing phenotypes. For example, lentiviral shRNA-mediated knockdown of ZRSR2 resulted in aberrant retention of U12-type introns similar to that observed in patient samples .
The specificity of ZRSR2's role in U12-type intron splicing is remarkable—in comparisons between ZRSR2 mutant and wild-type samples, 43 out of 45 introns consistently retained across all 64 comparison pairs were U12-dependent .
ZRSR2 loss has been associated with increased R-loop formation and DNA damage. To detect these phenomena:
R-loop detection:
DNA damage assessment:
Research has shown a small increase in γH2AX signals in ZRSR2-knockdown cells compared to control cells, suggesting that ZRSR2 deficiency promotes genomic instability . This methodology has helped establish a link between splicing defects and the DNA damage response pathway in MDS pathogenesis.
ZRSR2 deficiency appears to sensitize cells to inflammatory stimuli, particularly through the NF-κB pathway. To investigate this connection using ZRSR2 antibodies:
Pathway activation analysis: Treat ZRSR2 wild-type and knockdown cells with inflammatory stimuli (e.g., TNFα or LPS), then measure phosphorylated p65 (p-p65) levels using specific antibodies via Western blotting. Research has shown increased p-p65 levels in ZRSR2 knockdown cells following TNFα treatment, indicating enhanced NF-κB activation .
Protein-protein interaction studies: Use co-immunoprecipitation with ZRSR2 antibodies to identify interactions with components of inflammatory signaling pathways, potentially revealing direct molecular connections.
ChIP-seq analysis: Combine ZRSR2 antibodies with chromatin immunoprecipitation sequencing to map ZRSR2 binding sites at genes involved in inflammatory responses, potentially uncovering regulatory roles.
Transcriptional profiling: Compare inflammatory gene expression patterns between ZRSR2-deficient and normal cells using RNA-seq after cytokine stimulation, using ZRSR2 antibodies to validate knockdown efficiency.
This research direction is particularly relevant as activation of innate immune signaling has been proposed as a unifying mechanism mediating the effects of splicing factor mutations in MDS .
Optimizing ZRSR2 antibody usage requires technique-specific adjustments:
For Western Blotting:
Antibody dilution: Begin with 1:500-1:1000 for polyclonal antibodies
Blocking buffer: 5% non-fat milk or BSA in TBST
Incubation time: Overnight at 4°C for primary antibody
Detection method: Use enhanced chemiluminescence systems with appropriate sensitivity for low-abundance proteins like ZRSR2
Sample preparation: Include phosphatase inhibitors if studying phosphorylated forms
For Immunohistochemistry:
Antigen retrieval: Test both heat-mediated (citrate buffer) and enzymatic methods
Antibody dilution: Start with 1:100-1:200
Detection system: Use appropriate amplification systems (e.g., ABC, polymer-based)
Controls: Include both positive controls (tissues known to express ZRSR2) and negative controls
For Immunoprecipitation:
Pre-clearing samples to reduce non-specific binding
Cross-linking conditions when studying protein-RNA interactions
Buffer composition: Include RNase inhibitors when studying ZRSR2-RNA complexes
Elution conditions: Optimize to maintain protein activity
For all techniques:
Validate antibody specificity using ZRSR2 knockdown samples, as demonstrated in studies where lentiviral shRNA vectors efficiently downregulated ZRSR2 transcript and protein levels in 293T cells and leukemia cell lines (TF-1 and K562) .
To investigate ZRSR2's role in hematopoiesis:
Establish knockdown/knockout models:
Differentiation assays:
Culture hematopoietic stem/progenitor cells under conditions promoting differentiation
Monitor lineage markers over time by flow cytometry
Use ZRSR2 antibodies to correlate expression levels with differentiation stages
Cell proliferation and colony formation:
Compare colony-forming unit (CFU) capacity between ZRSR2-deficient and control cells
Measure proliferation using Ki-67 staining or BrdU incorporation
Intracellular signaling analysis:
In vivo models:
Transplant ZRSR2-deficient hematopoietic stem cells into irradiated recipients
Monitor engraftment, differentiation, and disease development
Use ZRSR2 antibodies for immunohistochemical analysis of bone marrow samples
When using ZRSR2 antibodies in splicing research, implement these controls and validation steps:
Antibody specificity controls:
Include ZRSR2 knockdown or knockout samples as negative controls
Test multiple antibodies targeting different epitopes of ZRSR2
Perform peptide competition assays to confirm specificity
Splicing functional controls:
Technical validation:
Confirm RNA-seq findings with RT-PCR for selected targets
Use multiple primer pairs spanning different exon-intron junctions
Validate protein-level consequences of splicing changes by Western blotting
Biological replicates:
Include sufficient biological replicates (≥3) to account for variability
For patient samples, stratify by ZRSR2 mutation status and clinical parameters
In the published research, eight ZRSR2 mutant MDS samples were compared with ZRSR2 wild-type MDS or normal bone marrow samples using rigorous statistical criteria (FDR<0.01, ΔMSI>20)
Quantification methods:
Use appropriate metrics for splicing efficiency (e.g., Mis-splicing Index)
Apply statistical thresholds (e.g., FDR<0.01) for identifying significant splicing events
Categorize introns as U12- or U2-type based on established sequence criteria
Synthetic lethality screens have identified potential therapeutic vulnerabilities in ZRSR2-deficient cells. To leverage these findings using antibody-based techniques:
Target validation in ZRSR2-deficient cells:
Western blotting with antibodies against potential synthetic lethal targets (e.g., ERBB3/4)
Immunoprecipitation to detect altered protein interactions in ZRSR2-deficient contexts
Screening results have identified increased sensitivity to ERBB3/4 knockdown and MAPK pathway inhibitors in ZRSR2-deficient cells
Pathway activation monitoring:
Use phospho-specific antibodies to monitor activation states of synthetic lethal pathways
Measure dynamic changes in response to inhibitor treatment
Compare pathway dynamics between ZRSR2 wild-type and deficient cells
Combinatorial treatment response assessment:
Detect markers of apoptosis, cell cycle arrest, or differentiation using specific antibodies following combination treatments
Monitor compensatory pathway activation that might indicate resistance mechanisms
In situ proximity ligation assays:
Detect protein-protein interactions between ZRSR2 (or its interacting partners) and components of synthetic lethal pathways
Visualize spatial relationships between proteins in intact cells
A systematic approach using these techniques can help translate initial screening findings into mechanistic understanding and therapeutic strategies for MDS patients with ZRSR2 mutations.
When analyzing patient samples with ZRSR2 antibodies, consider:
Sample preparation optimization:
For bone marrow biopsies, optimize fixation and antigen retrieval protocols
For peripheral blood, develop enrichment strategies for rare cell populations
Consider the impact of sample age and storage conditions on epitope integrity
Mutation-specific considerations:
Correlative analyses:
Compare ZRSR2 expression with clinical parameters (WHO subtype, IPSS score, cytogenetics)
Studies have shown ZRSR2 mutations are more prevalent in MDS subtypes without ring sideroblasts and in chronic myelomonocytic leukemia (CMML)
Correlate with markers of aberrant splicing or inflammatory activation
Reference standards:
Technical validation:
Confirm immunohistochemistry findings with orthogonal techniques (e.g., RNA-seq for splicing aberrations)
Use digital pathology tools for quantitative assessment of staining patterns
Patient data should include detailed clinical information as shown in the literature, where samples are characterized by age, gender, WHO subtype, IPSS score, and cytogenetic findings .
ZRSR2 contains serine/arginine-rich regions that are potential phosphorylation sites regulating its function. To assess ZRSR2 phosphorylation:
Phosphorylation-specific detection methods:
Use phospho-specific antibodies if available
Employ Phos-tag SDS-PAGE to separate phosphorylated from non-phosphorylated forms
Perform mass spectrometry analysis of immunoprecipitated ZRSR2 to map phosphorylation sites
Functional modulation experiments:
Context-dependent regulation:
Correlation with splicing activity:
Perform in vitro splicing assays with phosphorylated and dephosphorylated ZRSR2
Compare binding affinity to RNA and protein partners under different phosphorylation states
Understanding ZRSR2 phosphorylation may provide insights into its regulation and potential therapeutic approaches targeting its activity in disease contexts.
Several emerging technologies can expand ZRSR2 antibody applications:
Single-cell antibody-based technologies:
Combine ZRSR2 antibodies with single-cell RNA-seq to correlate protein levels with splicing patterns at single-cell resolution
Use mass cytometry (CyTOF) with ZRSR2 antibodies to analyze protein expression across heterogeneous cell populations
Implement proximity extension assays for ultrasensitive detection in limited sample material
Spatial biology approaches:
Apply multiplexed immunofluorescence to visualize ZRSR2 alongside other splicing factors in tissue context
Implement in situ sequencing to correlate ZRSR2 localization with splicing outcomes in intact tissues
Use spatial transcriptomics to map splicing patterns in relation to ZRSR2 expression
Dynamic interaction monitoring:
Employ FRET-based biosensors to track ZRSR2 interactions in living cells
Implement optogenetic approaches to modulate ZRSR2 activity with spatial and temporal precision
Use live-cell imaging with fluorescently tagged antibody fragments to track ZRSR2 dynamics
High-throughput functional screening:
Develop CRISPR activation/inhibition screens targeting ZRSR2 and interacting partners
Implement antibody-based readouts in high-content imaging screens
Research has already employed siRNA screens against the tyrosine kinome and small-molecule panels to identify synthetic lethal interactions with ZRSR2 deficiency
These technologies promise to advance our understanding of ZRSR2's role in normal and malignant hematopoiesis.
Integrating computational approaches with ZRSR2 antibody data can yield deeper insights:
Integrative multi-omics analysis:
Correlate ZRSR2 immunoprecipitation data with RNA-seq, CLIP-seq, and proteomics
Develop network models incorporating ZRSR2 protein interactions and splicing outcomes
Implement machine learning algorithms to predict the impact of ZRSR2 mutations on splicing patterns
Structural biology integration:
Use structural prediction tools to model ZRSR2-antibody binding interfaces
Simulate the impact of mutations on ZRSR2 protein structure and function
Design epitope-specific antibodies based on structural information
Splicing outcome prediction:
Develop algorithms to predict U12-type intron retention based on ZRSR2 expression levels
Create computational pipelines specifically optimized for minor intron detection
Studies have shown that introns can be categorized as U2- or U12-type based on the divergence at the 5' and 3' splice sites and the branchpoint sequence
Clinical data correlation:
Implement statistical methods to correlate ZRSR2 protein levels with clinical outcomes
Develop predictive models for treatment response based on ZRSR2 status
Create decision support tools for precision medicine approaches in MDS