ZBTB33 Antibody

Shipped with Ice Packs
In Stock

Description

Product Overview

ZBTB33 antibodies are polyclonal or monoclonal reagents designed to detect and analyze ZBTB33 protein expression across multiple experimental platforms. Key commercial products include:

Catalog NumberHostReactivityApplicationsSupplier
55270-1-APRabbitHumanWB, IHC, IF, IP, ChIP, ELISAProteintech
CAB12856RabbitHumanWBAssay Genie
ABIN1949419RabbitHuman, DogWB, IPAntibodies-Online
HPA005732RabbitHumanIF, IHCSigma-Aldrich

These antibodies are validated for specificity, with immunogens often derived from peptide sequences within ZBTB33’s functional domains (e.g., residues 375–425 or 564–673) .

Cell Cycle Regulation

ZBTB33 exhibits cell-type-specific roles in proliferation:

  • In HeLa cells, ZBTB33 promotes G1/S transition by binding promoters of cyclin D1 and cyclin E1, enhancing RB1 phosphorylation and E2F activity .

  • In HEK293 cells, ZBTB33 suppresses cyclin E1 expression, delaying G1 progression .

Cancer Biology

  • Breast Cancer: Subcellular localization of ZBTB33 correlates with LC3A/B expression, tumor immune microenvironment features, and patient survival .

  • Clonal Hematopoiesis: Somatic ZBTB33 mutations are linked to myelodysplastic syndromes (MDS) and confer competitive advantage in hematopoietic stem cells via altered RNA splicing .

DNA Methylation and Chromatin Remodeling

ZBTB33 binds methylated CpG islands at heterochromatin regions, regulating gene silencing through recruitment of NuRD/TRIM28 complexes .

Applications in Research

  • Western Blotting: Detects endogenous ZBTB33 at ~68–74 kDa in human cell lysates .

  • Immunohistochemistry: Localizes ZBTB33 in nuclear and cytoplasmic compartments of tumor tissues .

  • Functional Studies: siRNA-mediated depletion or overexpression reveals ZBTB33’s context-dependent roles in proliferation and differentiation .

Clinical and Mechanistic Insights

  • Prognostic Biomarker: ZBTB33 expression patterns in breast cancer correlate with autophagy markers (LC3A/B) and racial disparities in survival outcomes .

  • Epigenetic Crosstalk: ZBTB33 bridges DNA methylation and RNA splicing pathways, with mutations altering spliceosome interactions (e.g., SF3B1, SRSF2) .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery timelines may vary depending on the mode of purchase and location. For specific delivery details, please consult your local distributor.
Synonyms
Kaiso antibody; Kaiso protein antibody; Kaiso transcription factor antibody; KAISO_HUMAN antibody; Transcriptional regulator Kaiso antibody; ZBTB 33 antibody; Zbtb33 antibody; Zinc finger and BTB domain containing 33 antibody; Zinc finger and BTB domain-containing protein 33 antibody; Zinc finger transcription factor Kaiso antibody; ZNF 348 antibody; ZNF Kaiso antibody; ZNF348 antibody
Target Names
ZBTB33
Uniprot No.

Target Background

Function
ZBTB33, also known as Kaiso, is a transcriptional regulator exhibiting bimodal DNA-binding specificity. It binds to methylated CpG dinucleotides within the consensus sequence 5'-CGCG-3' and also interacts with the non-methylated consensus sequence 5'-CTGCNA-3', referred to as the consensus Kaiso binding site (KBS). Kaiso recruits the N-CoR repressor complex, facilitating histone deacetylation and the formation of repressive chromatin structures at target gene promoters. It potentially contributes to the repression of target genes involved in the Wnt signaling pathway. Kaiso can also activate transcription of a subset of target genes through the recruitment of CTNND2. Notably, Kaiso represses MMP7 expression in conjunction with transcriptional corepressors CBFA2T3, CBFA2T2, and RUNX1T1.
Gene References Into Functions
  1. miR-4262 expression was found to be significantly reduced in CC tissues and cell lines. Furthermore, decreased miR-4262 expression was strongly correlated with increased Kaiso (ZBTB33) expression. PMID: 28800784
  2. Extracellular microenvironment signals regulate RhoH and Kaiso, modulating actin-cytoskeleton structure and transcriptional activity during T cell migration. PMID: 27574848
  3. Kaiso silencing exhibited effects similar to miR-181a overexpression in glioma cells, while Kaiso overexpression in glioma cells partially reversed the inhibitory effects of the miR-181a mimic. PMID: 30036882
  4. Research indicates that Kaiso exhibits a preference for an mCpG site over a CpG site by two orders of magnitude in affinity, with a significant contribution from hydrophobic and CH...O interactions involving E535. PMID: 29546986
  5. Elevated nuclear Kaiso expression was observed in patients with a higher degree of African heritage compared to their Caucasian counterparts, suggesting a potential role for Kaiso in triple-negative breast cancer racial disparity. PMID: 28887687
  6. Kaiso promotes Jagged-1 expression, which may have implications in Notch-mediated colon cancer progression. PMID: 28637464
  7. Kaiso is implicated in cell proliferation. PMID: 28333150
  8. This research reports on the pathological characteristics of metastatic activity in parental (Kaiso-positive) versus Kaiso-depleted MDA-231 cells. PMID: 28880889
  9. The findings suggest a role for Kaiso in the progression of pancreatic ductal adenocarcinomas, involving the epithelial mesenchymal transformation markers, E-cadherin and Zeb1. PMID: 27424525
  10. This study reveals a novel mechanism by which ZBTB33 mediates the cyclin D1/cyclin E1/RB1/E2F pathway, controlling passage through the G1 restriction point and accelerating cancer cell proliferation. PMID: 27694442
  11. GR is a putative target gene of Kaiso. PMID: 26424557
  12. High Kaiso expression was correlated with invasion in prostate cancer. PMID: 26734997
  13. Results demonstrate that Kaiso binding to the unmethylated Kaiso binding site in the human ICR1 is essential for ICR1 methylation maintenance and influences transcription rates of the lncRNA H19. PMID: 27152123
  14. Kaiso modulates HIF1A gene expression by binding to the methylated HIF1A promoter in a region proximal to the autoregulatory HIF-1 binding site, primarily during hypoxia. PMID: 26514431
  15. The NF-kappaB response element, located near the p53RE#1, mediates APAF1 transcriptional repression by affecting the interaction between KAISO and p53. PMID: 26183023
  16. KAISO acts as a regulator of p53-mediated transcription of CDKN1A and apoptotic genes. PMID: 25288747
  17. Data suggest that Kaiso protein participates in the regulation of beta-catenin mRNA expression by interacting with p120-catenin in lung cancer cell lines. PMID: 24498333
  18. Nuclear localization of Kaiso promotes the poorly differentiated phenotype and EMT in infiltrating ductal carcinomas. PMID: 24570268
  19. Optimized knockdown with p120 and Kaiso siRNAs further expands the size of HCEC monolayers without endothelial mesenchymal transition (EMT) through selective activation of p120/Kaiso signaling, requiring the RhoA-ROCK-noncanonical BMP-NFkB signaling pathway. PMID: 24474278
  20. Transcription factor Kaiso does not interact with hydroxymethylated DNA within the CTGCNA sequence context. PMID: 23888785
  21. The presence of MTG16 in this complex, and its contributions to transcriptional repression, both necessitate Kaiso binding to its binding site on DNA, establishing MTG16-Kaiso binding as functionally relevant in Kaiso-dependent transcriptional repression. PMID: 23251453
  22. Kaiso represses the cell cycle gene cyclin D1 through a sequence-specific and methyl-CpG-dependent mechanism. PMID: 23226276
  23. These findings establish a defined oncogenic role for Kaiso in promoting the progression of prostate cancer. PMID: 22974583
  24. High cytoplasmic Kaiso expression is associated with thymoma. PMID: 22833212
  25. Nuclear Kaiso is prevalent in clinically aggressive ductal breast cancer, while cytoplasmic Kaiso and p120-mediated relief of Kaiso-dependent transcriptional repression characterize invasive lobular carcinoma (ILC). PMID: 22662240
  26. This study elucidates the molecular basis for recognition of methylated and specific DNA sequences by the zinc finger protein Kaiso. PMID: 22949637
  27. Selective activation of p120ctn-Kaiso signaling unlocks contact inhibition of ARPE-19 cells without epithelial-mesenchymal transition. PMID: 22590627
  28. Data demonstrate that Kaiso requires all three zinc fingers, along with adjacent protein regions, for DNA recognition. PMID: 22300642
  29. Results indicate that p120ctn isoforms 1 and 3 up-regulate cyclin D1, and consequently cyclin E, leading to the promotion of cell proliferation and cell cycle progression in lung cancer cells. PMID: 22276175
  30. Upon release from E-cadherin via Wnt3a-stimulated phosphorylation, p120-catenin regulates Kaiso activity, enhancing its binding to repressed promoters and relieving its inhibition of the beta-catenin-Tcf-4 transcriptional complex. PMID: 21670201
  31. Increased delta-catenin expression is crucial for maintaining the malignant phenotype of lung cancer, making delta-catenin a potential target protein for future cancer therapeutics. PMID: 21070476
  32. In endothelial cells, p120ctn exhibits a transcription repression function through the regulation of Kaiso, potentially acting as a cofactor with the transcription factor. PMID: 20382170
  33. Data strongly suggest that Kaiso's function as a transcriptional regulator may be linked to the control of the cell cycle and cell proliferation in cancer. PMID: 20169156
  34. This research investigates the bifunctional role of domain zinc fingers of the methyl-DNA-binding protein Kaiso. PMID: 20586187
  35. Differential expression of p120(ctn) and Kaiso mRNA was observed in human coronary artery endothelial cells depending on the application of laminar shear stress in relation to the wounding process. PMID: 14699141
  36. Kaiso translocates to the nucleus to regulate transcription of genes with diverse roles in cell growth and development. PMID: 15564377
  37. The findings imply an unexpected influence of the microenvironment on Kaiso expression and localization in colorectal adenocarcinoma. PMID: 15781635
  38. Kaiso associates with the matrilysin promoter in vivo. PMID: 15817151
  39. KAISO binds specifically to the methylated, but not the unmethylated, sequence in the first exon of the tyrosine hydroxylase [TH]gene and thus may play a significant role in modulating human TH gene expression. PMID: 15953356
  40. The Kaiso-CTCF interaction negatively regulates CTCF insulator activity. PMID: 16230345
  41. This study suggests that this gene is not implicated in the RTT molecular pathogenesis. PMID: 16530985
  42. p120 and Kaiso regulate Helicobacter pylori-induced expression of matrix metalloproteinase-7. PMID: 18653469
  43. Kaiso represses methylated tumor suppressor genes and can bind in a methylation-dependent manner to the CDKN2A in human colon cancer cell lines. Kaiso depletion induced tumor suppressor gene expression without affecting DNA methylation levels. PMID: 18794111
  44. Kaiso plays a specific role in centrosome assembly and function, extending beyond mitosis. PMID: 19502788
  45. Data suggest that cytoplasmic Kaiso expression is associated with poor prognosis of NSCLC, and various subcellular localizations of Kaiso may play differential biological roles in NSCLC. PMID: 19508730

Show More

Hide All

Database Links

HGNC: 16682

OMIM: 300329

KEGG: hsa:10009

STRING: 9606.ENSP00000314153

UniGene: Hs.143604

Subcellular Location
Nucleus. Cytoplasm. Note=Also cytoplasmic in cells grown at high densities.
Tissue Specificity
Expressed in vascular endothelium.

Q&A

What is ZBTB33 and why is it significant in research?

ZBTB33 (Zinc finger and BTB domain-containing protein 33), also known as Kaiso, is a unique transcription factor belonging to the zinc finger family of methyl-CpG-binding proteins. It has gained significance due to its bimodal DNA recognition capability, enabling it to target both methylated DNA and sequence-specific non-methylated DNA sites (called Kaiso-binding sites or KBS: TCCTGCNA) using the same set of three Cys2His2 zinc fingers . This dual binding capability allows ZBTB33 to function as both a transcriptional repressor and activator depending on cellular context, making it a fascinating subject for epigenetic regulation studies . Recent research has implicated ZBTB33 mutations in clonal hematopoiesis and myelodysplastic syndromes, highlighting its potential role in hematological disorders .

What are the known domains of ZBTB33 that antibodies typically target?

ZBTB33 contains several functional domains that are important for its biological activity and serve as potential epitopes for antibody development:

  • N-terminal BTB/POZ protein-protein interaction domain

  • Three C-terminal zinc finger (ZF) DNA-binding domains

  • Two regions necessary for ZBTB33's association with centrosomes and the mitotic spindle (SA1 and SA2)

  • Two sumo-interacting motifs (SIM)

Most commercially available antibodies target either the BTB domain or the zinc finger regions, as these are highly conserved and represent functional epitopes. When selecting an antibody, researchers should consider which domain might be masked in their specific experimental context due to protein-protein interactions.

How do ZBTB33 mutations affect antibody selection for research?

ZBTB33 mutations, particularly those observed in hematological disorders, cluster in functional domains including the BTB domain, zinc finger domains, centrosome association domains (SA1/SA2), and sumo-interacting motifs . When studying mutated forms of ZBTB33, researchers should:

  • Select antibodies whose epitopes lie outside the mutated region to ensure detection

  • Consider using multiple antibodies targeting different domains to compare expression patterns

  • Validate antibody specificity with positive and negative controls, particularly when studying patient samples with potential ZBTB33 mutations

  • For X-linked ZBTB33 mutations (which are more common in males), be aware that VAF (variant allele frequency) tends to be higher in males (mean 0.63) compared to females (mean 0.23)

What are the optimal applications for ZBTB33 antibodies in molecular biology research?

Based on validated research protocols, ZBTB33 antibodies have demonstrated effectiveness in several applications with specific methodological considerations:

ApplicationValidation StatusKey Methodological Considerations
Western Blotting (WB)Highly validatedOptimal with reducing conditions; ZBTB33 appears at ~100 kDa
Immunoprecipitation (IP)ValidatedEffective for protein-protein interaction studies, especially for splicing factors
Immunohistochemistry (IHC)Moderately validatedFixation-sensitive; paraformaldehyde preferred over methanol
Immunocytochemistry (ICC)ValidatedNuclear localization should be confirmed; can detect mitochondrial fraction
Immunofluorescence (IF)ValidatedShows primarily nuclear staining with some cytoplasmic signal
ChIP assaysLimited validationFunctional for detecting ZBTB33 binding to cyclin promoters

When designing experiments, researchers should consider that ZBTB33 can localize to both the nucleus (primary) and mitochondria (secondary), which has been confirmed through cellular fractionation studies .

How should researchers optimize chromatin immunoprecipitation (ChIP) protocols for ZBTB33?

Optimizing ChIP protocols for ZBTB33 requires special considerations due to its bimodal DNA binding characteristics:

  • Crosslinking optimization: Use dual crosslinking with both formaldehyde (1%) and ethylene glycol bis(succinimidyl succinate) (EGS, 2mM) to capture both protein-DNA and protein-protein interactions

  • Sonication parameters: Aim for 200-500bp fragments, as ZBTB33 binding sites can be proximity-dependent

  • Antibody selection: Choose antibodies validated specifically for ChIP applications that target the DNA-binding domain

  • Controls for bimodal binding:

    • Include methylated DNA controls (like known methylated promoters)

    • Include sequence-specific KBS site controls (TCCTGCNA motifs)

  • PCR primer design: For ZBTB33 targets like cyclin D1 and cyclin E1 promoters, design primers that flank both potential methylation sites and KBS sequences

When analyzing results, be aware that ZBTB33 occupies promoters in a context-dependent manner - for example, it binds cyclin D1 promoter via KBS sites and cyclin E1 promoter through methyl-specific interactions in HeLa cells .

What are the validated protocols for detecting endogenous versus exogenous ZBTB33?

Detection protocols differ significantly between endogenous and exogenous ZBTB33:

For endogenous ZBTB33:

  • Use sensitive detection methods as endogenous levels may be low in some cell types

  • Perform nuclear fractionation to concentrate the protein prior to analysis

  • Include RNase treatment in IP protocols to eliminate RNA-dependent interactions

  • For mitochondrial ZBTB33 detection, use mitochondrial fractionation with verification using mitochondrial markers like COX IV

For exogenous (tagged) ZBTB33:

  • When using V5-tagged constructs (as in published studies), validate expression using both anti-V5 and anti-ZBTB33 antibodies

  • Verify that tagged ZBTB33 maintains proper subcellular localization

  • For mutant ZBTB33 studies (e.g., R26C, G438D, C552R), confirm protein stability by immunoblotting as these mutations do not typically affect protein stability

How does ZBTB33 antibody reactivity differ across cell and tissue types?

ZBTB33 antibody reactivity exhibits significant cell-type variability due to:

  • Expression level differences: ZBTB33 expression varies substantially across cell types, with notably different levels between HeLa and HEK293 cells

  • Post-translational modifications: Cell-specific phosphorylation, SUMOylation, and other modifications can mask epitopes

  • Protein-protein interactions: ZBTB33 interacts with different partners in different cell types, potentially masking antibody binding sites

  • Subcellular localization differences:

    • In HeLa cells: Predominantly nuclear with functional activity at cell cycle gene promoters

    • In HEK293 cells: More distributed pattern with different functional outcomes

    • In hematopoietic cells: Both nuclear and mitochondrial fractions are detectable

Researchers should perform validation in their specific cell type of interest rather than relying solely on antibody validation data from different cellular contexts.

What controls are essential when using ZBTB33 antibodies in different experimental systems?

Essential controls for ZBTB33 antibody experiments include:

  • Genetic controls:

    • ZBTB33 knockdown/knockout cells (siRNA or CRISPR)

    • Rescue experiments with wild-type ZBTB33 expression

    • Expression of specific domain mutants (BTB, zinc finger, SA1/2, SIM)

  • Peptide competition controls:

    • Pre-incubation with immunizing peptide should abolish specific signals

  • Cell-type specific controls:

    • Parallel experiments in cell lines with known ZBTB33 function (HeLa vs. HEK293)

    • Include both positive (high expression) and negative (low expression) cell types

  • Application-specific controls:

    • For IP: IgG control pulldowns

    • For ChIP: Input DNA, IgG ChIP, and non-target loci

    • For IHC/ICC: Secondary antibody only controls

  • Domain-specific controls:

    • When studying mutant ZBTB33 (e.g., R26C), include wild-type controls alongside mutant samples

How can researchers differentiate between ZBTB33's dual DNA binding modalities using antibodies?

Differentiating between ZBTB33's methylated DNA binding and sequence-specific (KBS) binding requires specialized approaches:

  • Combined ChIP and bisulfite sequencing:

    • Perform ChIP with validated ZBTB33 antibodies

    • Analyze immunoprecipitated DNA with bisulfite sequencing to determine methylation status

    • Compare with motif analysis for KBS sites (TCCTGCNA)

  • Mutant ZBTB33 studies:

    • Generate mutations in zinc finger domains that selectively disrupt either methylated DNA binding or KBS binding

    • Use antibodies against tagged mutant proteins to determine binding specificity

  • Competitive binding assays:

    • Pre-incubate nuclear extracts with methylated or unmethylated DNA competitors

    • Perform ChIP to determine which type of binding is affected

  • Validation approach using cyclin promoters:

    • As demonstrated in research, ZBTB33 binds cyclin D1 promoter via KBS and cyclin E1 via methylated DNA

    • These can serve as positive controls for distinguishing binding modalities

What strategies can resolve antibody cross-reactivity with other ZBTB family members?

ZBTB33 belongs to a family that includes ZBTB4 and ZBTB38, which share structural similarities that can lead to cross-reactivity. To resolve this:

  • Epitope selection:

    • Choose antibodies targeting unique regions with minimal sequence homology to ZBTB4/ZBTB38

    • Avoid antibodies targeting the highly conserved zinc finger domains when possible

  • Validation approaches:

    • Test antibody specificity in cells with ZBTB33 knockdown/knockout

    • Perform parallel testing in cells expressing predominantly one family member

    • Use recombinant protein competition with all three family members

  • Sequential immunoprecipitation:

    • Deplete extracts of ZBTB4/ZBTB38 using specific antibodies before ZBTB33 IP

    • Confirm specificity via mass spectrometry analysis of immunoprecipitates

  • Size-based discrimination:

    • Leverage size differences between family members (ZBTB33: ~100kDa; ZBTB4: ~119kDa; ZBTB38: ~132kDa)

    • Use high-resolution SDS-PAGE to clearly separate proteins by molecular weight

How can researchers effectively use ZBTB33 antibodies in studying hematopoietic stem cell biology?

Based on recent findings linking ZBTB33 mutations to clonal hematopoiesis and myelodysplastic syndromes , specialized approaches for hematopoietic research include:

  • Flow cytometry applications:

    • Optimize fixation/permeabilization for nuclear ZBTB33 detection

    • Combine with stem cell markers (CD34, CD38, CD90, etc.) for population-specific analysis

  • Mutation-specific detection:

    • When studying patient samples, complement antibody studies with sequencing

    • Use phospho-specific antibodies to monitor downstream signaling effects

  • Transplantation experiment design:

    • For mouse models with ZBTB33 mutations (as used in published studies), track donor cell engraftment with concurrent antibody staining

    • Monitor competitive advantage of ZBTB33-edited cells using flow cytometry

  • RNA splicing analysis:

    • Since ZBTB33 interacts with splicing factors in hematopoietic cells, combine IP with RNA-seq

    • Focus on intron retention events, which are significantly affected by ZBTB33 mutations

  • HSPC isolation protocols:

    • When isolating LSK (Lin-Sca+Kit+) cells, use careful fixation to preserve epitopes

    • For rare hematopoietic populations, consider proximity ligation assays instead of traditional IF for enhanced sensitivity

How should researchers interpret contradictory ZBTB33 antibody results between different cell types?

ZBTB33 exhibits cell-type specific functions that can lead to seemingly contradictory results. Research has shown that ZBTB33:

  • Acts as a pro-proliferative factor in HeLa cells but anti-proliferative in HEK293 cells

  • Shows different subcellular distributions depending on cell context

  • Interacts with different protein partners in different cell types

When faced with contradictory results:

  • Verify antibody specificity in each cell type independently

  • Consider that ZBTB33 can regulate the same genes differently in different contexts

  • Examine the cell-specific transcriptome to identify unique binding partners

  • Assess the methylation status of target genes in each cell type

  • Conduct parallel RNA-seq studies to identify cell-specific ZBTB33-regulated genes

The published contradictory roles in cell cycle regulation between HeLa and HEK293 cells provide a framework for understanding such discrepancies - in HeLa cells, ZBTB33 accelerates G1-S transition by enhancing cyclin expression, while in HEK293 cells it decelerates this transition .

What are the key considerations when analyzing ZBTB33 mutations in patient samples using antibodies?

When using antibodies to study ZBTB33 in patient samples with potential mutations:

  • Mutation distribution patterns:

    • ZBTB33 mutations cluster in functional domains (BTB, ZF, SA1/2, SIM)

    • Most mutations in clonal hematopoiesis and MDS are missense mutations

    • X-linked gene mutations show gender bias (more common in males)

  • Epitope accessibility issues:

    • Select antibodies whose epitopes are preserved in the expected mutation profile

    • Consider using multiple antibodies targeting different domains

  • Allele-specific detection:

    • For common mutations (e.g., R26C in BTB domain), consider developing mutation-specific antibodies

    • Complement antibody studies with genomic analysis (VAF determination)

  • Functional readouts:

    • Assess downstream effects on splicing (increased intron retention)

    • Monitor cell cycle progression markers in conjunction with ZBTB33 detection

  • Clinical correlation:

    • When analyzing patient samples, correlate ZBTB33 antibody staining patterns with:

      • Mutation status determined by sequencing

      • Clinical parameters

      • Response to treatments

How can researchers use ZBTB33 antibodies to investigate its interaction with the RNA splicing machinery?

Recent research has revealed ZBTB33 interactions with splicing-associated proteins . To investigate these interactions:

  • Co-immunoprecipitation approaches:

    • Use ZBTB33 antibodies for pulldown followed by probing for splicing factors

    • Perform reverse IPs with antibodies against splicing factors (validated interactions include multiple RNA splicing proteins)

    • Include RNase treatment controls to distinguish RNA-dependent interactions

  • Proximity ligation assays:

    • Visualize in situ interactions between ZBTB33 and splicing factors

    • Quantify interaction differences between wild-type and mutant cells

  • RNA splicing analysis:

    • Compare splicing patterns (particularly intron retention) between:

      • ZBTB33 wild-type and knockout/knockdown cells

      • ZBTB33 wild-type and mutant (e.g., R26C) expressing cells

    • Focus on genes showing differential splicing following ZBTB33 depletion

  • Domain mapping:

    • Use truncated ZBTB33 constructs to determine which domains interact with splicing factors

    • Focus on the BTB domain, as the R26C mutation affects interaction with splicing proteins

This approach has revealed that ZBTB33-edited mouse hematopoietic stem cells exhibit increased genome-wide intron retention, connecting ZBTB33 function to RNA processing pathways .

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 2025 TheBiotek. All Rights Reserved.