IKZF1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
CLL associated antigen KW 6 antibody; DNA-binding protein Ikaros antibody; hIk 1 antibody; Hs.54452 antibody; IK1 antibody; Ikaros (zinc finger protein) antibody; IKAROS antibody; IKAROS family zinc finger 1 (Ikaros) antibody; Ikaros family zinc finger protein 1 antibody; Ikzf1 antibody; IKZF1_HUMAN antibody; LYF1 antibody; Lymphoid transcription factor LyF-1 antibody; PRO0758 antibody; Zinc finger protein subfamily 1A 1 (Ikaros) antibody; Zinc finger protein subfamily 1A 1 antibody; Zinc finger protein; subfamily 1A; member 1 antibody; ZNFN1A1 antibody
Target Names
Uniprot No.

Target Background

Function
IKZF1, also known as Ikaros, is a transcription regulator that plays a crucial role in hematopoietic cell differentiation. It binds to gamma-satellite DNA and is involved in the development of lymphocytes, including B- and T-cells. IKZF1 binds and activates the enhancer (delta-A element) of the CD3-delta gene. It acts as a repressor of the TDT (terminal deoxynucleotidyltransferase) gene during thymocyte differentiation. IKZF1 regulates transcription by associating with both HDAC-dependent and HDAC-independent complexes. It targets two chromatin-remodeling complexes, NuRD and BAF (SWI/SNF), in a single complex (PYR complex), to the beta-globin locus in adult erythrocytes. IKZF1 promotes normal apoptosis in adult erythroid cells and contributes to the early temporal competence of retinal progenitor cells (RPCs). Its function is isoform-specific and can be modulated by dominant-negative inactive isoforms.
Gene References Into Functions
  1. The cumulative recurrence rate of children with non-functional subtypes of IKZF1 was significantly higher than those with functional types of IKZF1. PMID: 30022754
  2. Heterozygous mutations in IKZF1 decrease plasmacytoid dendritic cell numbers and expand conventional dendritic cells. PMID: 29588478
  3. IKZF1 plays a role in childhood B-cell precursor acute lymphoblastic leukemia. PMID: 28947432
  4. Studies demonstrate that alterations in Ikaros expression promote B-1 cell differentiation into phagocytes. PMID: 29107383
  5. Results show that IKAROS promotes PHF2 expression, suggesting that low PHF2 expression, along with IKAROS gene deletion, drives oncogenesis of ALL. PMID: 28994305
  6. This case provides the first definitive evidence of an IKAROS heterozygous mutation causing both immunodeficiency and NOTCH1-driven T-ALL in humans. PMID: 28096536
  7. A review summarizes the regulation of leukemia by Casein kinase II, glycogen synthase kinase-3, and Ikaros. PMID: 28623166
  8. Research found Ikaros directly binds the DNM2 promoter and suppresses DNM2 expression in ALL tumors and cell lines. PMID: 27885263
  9. A clear distinction exists between loss-of-function and dominant-negative IKZF1 deletions. Patients with these deletions should be closely monitored for minimal residual disease to detect early relapses and may be candidates for alternative or intensified treatment regimens. PMID: 28751559
  10. IKZF1 gene deletion is associated with acute lymphoblastic leukemia. PMID: 27588474
  11. Data reveal the mechanism by which chromatin remodeling and target gene expression are regulated by Ikaros alone and in complex with HDAC1 in B-ALL. PMID: 26639180
  12. Ikaros regulates expression of the BCL6/BACH2 axis in acute lymphoblastic leukemia cells. PMID: 28030830
  13. High CRLF2 expression, alongside IKZF1 deletion, drives oncogenesis of acute lymphoblastic leukemia. PMID: 27391346
  14. Expression of both CEBPE and IKZF1 in patient leukemic B cells is most similar to that in early stage B cells, believed to be the cell-of-origin for B-ALL. PMID: 26437776
  15. IKZF1 is identified as a novel regulator of glucocorticoid-induced transcriptional responses and a critical determinant of glucocorticoid-mediated cell death in normal and leukemic B cells. PMID: 26713593
  16. A systematic screen of 6 potentially functional SNPs in ARID5B and IKZF1 genes was conducted. PMID: 29292192
  17. Two families exhibit a Common variable immunodeficiency-like syndrome with normal hematologic parameters and fetal hemoglobin silencing, associated with heterozygous IKZF1 mutations. PMID: 27581358
  18. The IKZF1 rs10235796 C allele, IKZF1 rs6964969A>G, CDKN2A rs3731246 G>C, and CDKN2A rs3731246 C allele were significantly associated with Acute Lymphoblastic Leukemia in Yemenis of Arab-Asian descent. A borderline association was found with the IKZF1 rs4132601 T>G variant. No associations were found with IKZF1 rs11978267 or rs7789635, DDC rs3779084; rs880028; rs7809758, CDKN2A rs3731217, CEBPE rs2239633; rs12434881. PMID: 28768142
  19. Silence of IKZF1 expression in MHCC-LM3 and MHCC-97L cell lines revealed a 1.84- and 2.27-fold rise in MDIG mRNA levels, respectively. PMID: 28471446
  20. Data show that six patients had large interstitial deletions starting within intronic regions of COBL at diagnosis, which is ~611 Kb downstream of IKZF1, suggesting that COBL is a hotspot for IKZF1 deletion (DeltaIKZF1). PMID: 27419633
  21. This study provides the first evidence for the association of IKZF1 variants with diffuse large B-cell lymphoma outcome. PMID: 28879630
  22. PTEN induced miR-26b expression by regulating the differential expression of Ikaros isoforms, which are transcriptional regulators of miR-26b. PMID: 28280276
  23. The mechanism of action of lenalidomide in ABC-DLBCL cells involves downregulation of SPIB transcription through cereblon-induced degradation of IKAROS, but not IKZF3. Small hairpin RNA-mediated knockdown of MYC enhanced the cytotoxicity of IMiDs. PMID: 28893618
  24. IKZF1 and IKZF3 expressions were associated with longer median progression-free survival and overall survival in multiple myeloma patients. PMID: 27881177
  25. Phosphorylation of Ikaros by CK2 impairs its DNA-binding ability, as well as its ability to regulate gene expression and function as a tumor suppressor in leukemia. (Review) PMID: 27666503
  26. High IKZF1 expression is associated with multiple myeloma. PMID: 28017969
  27. Bioinformatics analysis indicated that both SNPs were located in a putative enhancer area in immune-related cell lines and tissues. Protein-protein interaction analysis found that IKZF1, together with GTF2I (an SS susceptibility gene identified through GWAS), could interact with histone deacetylase family proteins. This is the first study to report an association between IKZF1 and SS in Han Chinese. PMID: 28552951
  28. The M4 motif (ACTAYRNNNCCCR) is a functional regulatory bipartite cis-element, which engages a THAP11/HCF-1 complex via binding to the ACTAYR module, while the CCCRRNRNRC subsequence part constitutes a binding platform for Ikaros and NFKB1. PMID: 27576892
  29. Germline heterozygous IKZF1 mutations cause dysgammaglobulinemia, hematologic abnormalities (including B-cell defects), and autoimmune diseases. PMID: 27939403
  30. Results suggest that the rs1456896 A allele is associated with protective susceptibility to lupus nephritis. However, this association did not seem to be implicated in the disease and histopathological severity of lupus nephritis in a northern Han Chinese cohort. PMID: 27684961
  31. Elevated Ctnnd1 expression contributes to the maintenance of murine B-ALL cells with compromised Ikaros function. PMID: 28190000
  32. The presence of IKAROS deletion in acute lymphoblastic leukemia in Mexican Mestizos patients could represent a poor-prognosis marker and was probably related to therapy failure. PMID: 27623040
  33. Sumoylated Ikaros is less effective than unsumoylated forms at inhibiting the expansion of murine leukemic cells. Ikaros sumoylation is abundant in human B-cell acute lymphoblastic leukemic cells, but not in healthy peripheral blood leukocytes. These findings suggest that sumoylation may be important in modulating the tumor suppressor function of Ikaros. PMID: 27315244
  34. BTG1 is identified as a tumor suppressor in leukemia. Deletion of BTG1 strongly enhances the risk of relapse in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia and augments the glucocorticoid resistance phenotype mediated by the loss of IKZF1 function. PMID: 27979924
  35. ETV6 and IKZF1 are transcriptional regulators not just of ERG, but also of a number of genes regulated by a densely interconnected network of seven transcription factors. PMID: 27604872
  36. CRBN binds to Ikaros via its N-terminal region and regulates transcriptional activities of Ikaros and its downstream target, enkephalin. PMID: 27329811
  37. Research revealed over 170 NFAT-associated proteins, half of which are involved in transcriptional regulation. Among them are many hitherto unknown interaction partners of NFATc1 and NFATc2 in T cells, such as Raptor, CHEK1, CREB1, RUNX1, SATB1, Ikaros, and Helios. PMID: 27637333
  38. Ikaros undergoes a transient increase in protein levels at the transitional single-positive CD8+ developmental stage before diverging in their expression patterns at later stages. PMID: 27502439
  39. Variants within IKZF1, ARID5B, and CEBPE were associated with pediatric ALL risks. PMID: 27184773
  40. FISH studies showed false-negative results in 10%, 40%, and 28% of the samples tested for the IKZF1, PAX5, and CDKN2A/B gene deletions, respectively. The PAX5 and IKZF1 abnormalities are highly specific to B-ALL and can be used as diagnostic markers. PMID: 28214896
  41. The impact of IKZF1 polymorphisms on childhood ALL risk was investigated. PMID: 26790447
  42. Genotypic and allelic frequencies differed significantly between cases and controls at IKZF1-rs4132601 (p=0.039, p=0.015) and ARID5B-rs10821936 (p=0.028, p=0.026). PMID: 27644650
  43. Data indicate that IKZF1 deletion may be a useful prognostic variable in adults with common B-cell ALL, particularly in persons without BCR-ABL1 and those receiving chemotherapy-only. PMID: 27067989
  44. Research suggests an oncogenic role for an Ikaros protein/MYCBP2 protein/proto-oncogene protein c-MYC axis in adult acute lymphoblastic leukemia (ALL), providing a mechanism for target therapies that activate Ikaros in ALL. PMID: 26517351
  45. The anti-PEL effects of IMiDs involved cereblon-dependent suppression of IRF4 and rapid degradation of IKZF1, but not IKZF3. Small hairpin RNA-mediated knockdown of MYC enhanced the cytotoxicity of IMiDs. PMID: 26119939
  46. A novel, non-canonical splice variant of the Ikaros gene (Ik11) is aberrantly expressed in B-cell lymphoproliferative disorders. PMID: 23874502
  47. Data suggest a mechanism through which Ikaros and HDAC1 regulate the epigenetic signature in leukemia: via regulation of JARID1B transcription. PMID: 26655717
  48. IKZF1 deletions were detected in 28.7% of B-ALL patients, were more common in BCR-ABL positive and adult B-ALL, and correlated with higher induction failure. PMID: 26704074
  49. All variants of rare IKZF1 deletions are associated with an unfavorable prognosis in pediatric BCP-ALL. PMID: 26202931
  50. A genome-wide association study for Cold Medicine related -Stevens-Johnson Syndrome /Toxic epidermal necrolysis with SOCs found that IKZF1 single-nucleotide polymorphisms (SNPs) were significantly associated. PMID: 26448174

Show More

Hide All

Database Links

HGNC: 13176

OMIM: 603023

KEGG: hsa:10320

STRING: 9606.ENSP00000331614

UniGene: Hs.435949

Involvement In Disease
Immunodeficiency, common variable, 13 (CVID13)
Protein Families
Ikaros C2H2-type zinc-finger protein family
Subcellular Location
Nucleus.; [Isoform Ik2]: Nucleus.; [Isoform Ik6]: Cytoplasm.
Tissue Specificity
Abundantly expressed in thymus, spleen and peripheral blood Leukocytes and lymph nodes. Lower expression in bone marrow and small intestine.

Q&A

What is IKZF1 and why is it significant in research?

IKZF1 (IKAROS family zinc finger 1) is a critical transcription factor encoded by the IKZF1 gene in humans. Also known as IKAROS or CVID13, this protein has a molecular weight of approximately 57.5 kilodaltons. IKZF1 plays a crucial role in hematopoiesis, particularly in the development and differentiation of lymphoid cells. Its significance in research stems from its involvement in various immunological disorders and hematological malignancies, making it an important target for understanding disease mechanisms and developing therapeutic strategies . It functions as a master transcription factor with crucial roles at multiple stages of lymphocyte development and hematopoiesis .

What species reactivity should be considered when selecting an IKZF1 antibody?

When selecting an IKZF1 antibody, researchers should consider species reactivity based on their experimental model. Commercial IKZF1 antibodies are available with reactivity against human, mouse, and rat orthologs, with some also recognizing canine, porcine, and monkey IKZF1. Cross-reactivity between species varies between antibody clones, so researchers should verify the specific reactivity profile when selecting an antibody for multi-species studies . For studies involving transgenic or xenograft models, selecting antibodies that can distinguish between human and host species IKZF1 is essential for proper data interpretation.

What are the common applications for IKZF1 antibodies in research?

IKZF1 antibodies are used in multiple experimental applications including:

ApplicationCommon DilutionsKey Considerations
Western Blot (WB)1:500-1:2000Effective for detecting native and denatured IKZF1
Immunofluorescence (IF)1:50-1:200Allows subcellular localization studies
Immunohistochemistry (IHC)1:50-1:200For tissue section analysis, often with paraffin samples
Flow Cytometry (FCM)1:50-1:100For cell population analysis and sorting
Chromatin Immunoprecipitation (ChIP)1:50-1:100For studying DNA-protein interactions

The selection of application should be guided by experimental goals, with verification of the antibody's validation for specific applications prior to use . For multi-parameter studies, conjugated antibodies (FITC, APC, etc.) may be preferable, while unconjugated antibodies offer greater flexibility with secondary detection systems.

How do IKZF1 alterations impact drug sensitivity in B-cell acute lymphoblastic leukemia models?

IKZF1 alterations significantly modify chemosensitivity profiles in B-ALL models. Research using CRISPR/Cas9-engineered cell lines with various IKZF1 lesions has demonstrated that IKZF1 knockout and heterozygous null cells exhibit resistance to multiple common B-ALL therapies, including dexamethasone, asparaginase, and daunorubicin. The mechanism appears to involve a shift toward a stem/myeloid cell-like phenotype with JAK/STAT pathway upregulation .

Interestingly, IKZF1 knockout cells show increased sensitivity to cytarabine due to downregulation of SAMHD1, which can be reversed through SAMHD1 restoration. This differential drug sensitivity profile suggests that precise characterization of IKZF1 status could enhance therapeutic stratification for B-ALL patients . When designing experiments to investigate IKZF1-related drug resistance, researchers should include multiple drug classes and consider downstream effectors like SAMHD1 that mediate specific sensitivity patterns.

What phenotypic differences exist between IKZF1 knockout models versus dominant-negative IK6 isoform expression?

IKZF1 knockout and dominant-negative IK6 isoform models exhibit both shared and distinct phenotypes:

ParameterIKZF1 KnockoutIK6 Dominant-Negative
Drug ResistanceResistant to dexamethasone, asparaginase, daunorubicinSimilar resistance profile but retains dexamethasone sensitivity
Cytarabine SensitivityIncreasedIncreased
In vivo EngraftmentEnhancedEnhanced
Circulating BlastsIncreasedIncreased
Organ InfiltrationIncreasedIncreased
Survival TimeDecreasedDecreased

These differences highlight the importance of precise characterization of IKZF1 alterations in both research and clinical settings . Researchers investigating IKZF1 alterations should consider generating both knockout and dominant-negative models to comprehensively understand the spectrum of functional consequences.

How do gain-of-function IKZF1 variants differ from loss-of-function variants in immune dysregulation?

Gain-of-function (GOF) IKZF1 variants (particularly R183C/H) manifest distinctly different phenotypes compared to haploinsufficient (HI) or dominant-negative (DN) variants:

ParameterGOF IKZF1 Variants (R183C/H)Loss-of-function Variants
Clinical PresentationInflammatory, autoimmune, allergic symptoms, abnormal plasma cell proliferationB cell immune deficiency, combined immunodeficiency
T Cell ProductionImpaired IL-2 productionVariable T cell defects
T Helper Cell BiasTH2 skewingLess pronounced helper bias
Regulatory T CellsReduced numbersVariable impact
EosinophiliaPresentNot typically observed
DNA BindingIncreasedDecreased
Response to LenalidomideTherapeutic potential through IKZF1 degradationLimited therapeutic value

These distinct profiles highlight the mechanistic complexity of IKZF1 in immune regulation and the importance of variant-specific characterization in both research and clinical settings . When designing studies investigating IKZF1 variants, researchers should incorporate comprehensive immune phenotyping to capture the full spectrum of cellular abnormalities.

What are optimal protocols for engineering IKZF1 mutations using CRISPR/Cas9?

Engineering IKZF1 mutations requires careful design considerations for successful outcomes:

  • Guide RNA Selection: Target sequences with minimal off-target effects, preferably in early exons (exons 3-5) for complete loss-of-function. For specific mutations like R183C/H, use homology-directed repair with appropriate donor templates.

  • Cell Type Considerations: For lymphoid studies, efficiency varies between cell types. Reported transfection efficiencies:

    • Pre-B ALL cell lines: 30-70% (electroporation)

    • Primary B cells: 15-30% (nucleofection)

    • T cell lines: 40-60% (electroporation)

  • Screening Strategy: Implement a tiered approach:

    • Initial screening: T7 endonuclease assay or heteroduplex mobility assay

    • Targeted sequencing for mutation confirmation

    • Western blotting to verify protein loss/alteration

    • Functional validation using IKZF1-dependent reporter systems

  • Clone Validation: Comprehensive validation should include sequencing, Western blot for protein expression, and at least one functional assay (e.g., known target gene expression analysis) .

When engineering specific mutations like IK6 isoform, homology-directed repair strategies with appropriate donor templates are essential for precise genetic modifications.

How can researchers optimize ChIP-seq experiments for IKZF1 binding studies?

Optimizing ChIP-seq for IKZF1 binding studies involves several critical considerations:

  • Antibody Selection: Use ChIP-validated antibodies targeting conserved C-terminal regions for full-length IKZF1 detection or N-terminal regions for detecting all isoforms. Monoclonal antibodies generally provide more consistent results across experiments.

  • Crosslinking Conditions:

    • Standard protocol: 1% formaldehyde for 10 minutes at room temperature

    • For detection of weaker/transient interactions: Use dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde

  • Sonication Parameters:

    • Target fragment size: 200-400bp

    • Typical conditions: 20-30 cycles of 30s ON/30s OFF at medium power

    • Critical quality control: Verify fragment size distribution using Bioanalyzer

  • Data Analysis Pipeline:

    • Peak calling: MACS2 with q-value < 0.05

    • Motif analysis: HOMER or MEME-ChIP for de novo motif discovery

    • Composite element identification: SPICE pipeline for detecting IKZF1 with partner factors (e.g., JUN)

  • Validation: Confirm key binding sites with ChIP-qPCR using independent biological replicates and correlate with gene expression data.

This comprehensive approach ensures high-quality IKZF1 binding data necessary for understanding its genomic interactions and transcriptional regulatory networks .

What are the most effective methods for detecting IKZF1 isoforms in research samples?

Detecting IKZF1 isoforms requires a multi-method approach for comprehensive characterization:

MethodAdvantagesLimitationsResolution
Western BlotDetects multiple isoforms simultaneouslySemi-quantitative; antibody-dependentModerate
RT-PCRHigh sensitivity for known variantsMay miss novel splice variantsHigh for known variants
Targeted RNA-seqComprehensive detection of splice variantsHigher cost; bioinformatic complexityVery high
Flow CytometrySingle-cell resolutionLimited to isoform-specific antibodiesModerate
Mass SpectrometryDirect protein detection; quantitativeSample preparation complexity; costVery high

For optimal sensitivity in detecting the dominant-negative IK6 isoform, a nested PCR approach using primers spanning exons 2-8 is recommended. Analysis of patient samples typically benefits from combining protein-level (Western blot) and transcript-level (RT-PCR) detection methods to comprehensively characterize IKZF1 status . When developing multiplexed assays, researchers should validate the specificity of each component using well-characterized control samples expressing different IKZF1 isoforms.

How does IKZF1 interact with JUN in composite elements, and what are the functional implications?

IKZF1 and JUN form a previously unrecognized composite element at regulatory regions, with important functional implications:

  • Interaction Characteristics:

    • Co-localization: ChIP-seq data shows extensive co-localization of IKZF1 and JUN in the human cell line GM12878 at multiple genomic loci

    • Structural basis: The interaction likely involves specific DNA motif spacing preferences identified through the SPICE (Spacing Preference Identification of Composite Elements) pipeline

    • Complex formation: JUN (as part of AP-1 complexes) and IKZF1 bind cooperatively, enhancing regulatory effects

  • Functional Implications:

    • Transcriptional regulation: Cooperative binding likely augments gene transcription at co-regulated loci

    • Immune cell development: Since both factors play critical roles in immune cells (JUN in T cells, IKZF1 in B and T cells), their interaction suggests coordinated regulation of immune development

    • Potential therapeutic target: Disrupting this interaction could have therapeutic implications in contexts where aberrant IKZF1 or JUN activity contributes to disease

Future research directions should explore how this interaction is affected by IKZF1 mutations in leukemia and immune disorders to determine its role in disease pathogenesis.

What are the downstream consequences of SAMHD1 downregulation after IKZF1 knockout?

SAMHD1 downregulation following IKZF1 knockout leads to several significant downstream consequences:

  • Enhanced Cytarabine Sensitivity:

    • SAMHD1 normally hydrolyzes the active metabolite of cytarabine (ara-CTP)

    • Decreased SAMHD1 results in higher ara-CTP levels and increased cytotoxicity

    • This creates a potential therapeutic vulnerability in IKZF1-altered leukemias

  • Altered DNA Damage Response:

    • SAMHD1 participates in DNA repair mechanisms

    • Downregulation may impair DNA damage response pathways

    • This potentially contributes to genomic instability

  • Innate Immune Modulation:

    • SAMHD1 regulates intracellular dNTP pools and restricts viral replication

    • Downregulation may alter cellular responses to viral infections

    • This creates potential immune surveillance defects

This mechanistic relationship provides a rational basis for exploring cytarabine-based therapeutic approaches in leukemias with IKZF1 alterations, particularly in combination with agents that inhibit compensatory survival pathways.

How does IKZF1 status influence JAK/STAT signaling in hematologic malignancies?

IKZF1 status significantly impacts JAK/STAT signaling through multiple mechanisms:

  • Transcriptional Regulation:

    • IKZF1 loss leads to upregulation of JAK/STAT pathway components

    • Transcriptome analysis reveals a stem/myeloid cell-like signature with JAK/STAT pathway activation

    • This creates potential dependencies on JAK/STAT signaling for survival

  • Clinical Implications:

    • In Philadelphia chromosome-positive (Ph+) leukemias, IKZF1 deletion may synergize with BCR-ABL to enhance JAK/STAT signaling

    • In Ph-like leukemias, IKZF1 alterations likely cooperate with other lesions activating JAK/STAT pathways

    • These interactions suggest potential therapeutic vulnerability to JAK inhibitors

  • Therapeutic Targeting:

    • JAK inhibitors (e.g., ruxolitinib) may have enhanced efficacy in IKZF1-altered leukemias

    • Combination approaches targeting both IKZF1 degradation (e.g., lenalidomide) and JAK/STAT inhibition represent a rational therapeutic strategy

When designing experiments to investigate these interactions, researchers should incorporate comprehensive phosphoprotein analysis to fully characterize the impact of IKZF1 status on JAK/STAT signaling dynamics.

What controls should be included when validating IKZF1 antibodies for specific applications?

Comprehensive validation of IKZF1 antibodies requires application-specific controls:

ApplicationPositive ControlsNegative ControlsValidation Criteria
Western BlotCell lines with known IKZF1 expression (NALM-6, Jurkat)IKZF1 knockout cell lines, siRNA knockdown samplesSingle band at 57.5 kDa; additional bands for known isoforms
IHC/IFLymphoid tissues (tonsil, spleen); known IKZF1-expressing cell linesIKZF1 knockout tissues/cells; isotype controlsNuclear staining pattern in lymphoid cells
Flow CytometryFresh PBMCs (B and T cells); IKZF1-expressing cell linesIKZF1 knockout cells; isotype controls; blocking peptide competitionDistinct population separation with expected expression patterns
ChIPLymphoid cell lines with verified IKZF1 binding sitesIKZF1 knockout cells; IgG control; non-target regionsEnrichment at known target genes vs. control regions

Additional validation approaches include:

  • Cross-validation with multiple antibodies targeting different epitopes

  • Correlation with mRNA expression data

  • Peptide competition assays to confirm specificity

  • Verification with recombinant IKZF1 protein standards

Thorough validation ensures experimental reliability and reproducibility, particularly in studies examining specific IKZF1 isoforms or mutant variants.

How should researchers interpret contradictory data between IKZF1 genomic, transcript, and protein analyses?

Resolving contradictions between IKZF1 genomic, transcript, and protein analyses requires systematic investigation:

  • Common Causes of Discrepancies:

    • Alternative splicing: Multiple IKZF1 isoforms may not be detected by all methods

    • Post-translational modifications: Affect protein detection but not genomic/transcript analysis

    • Antibody specificity: Some antibodies may not recognize all isoforms or may be affected by modifications

    • Technical artifacts: Sample processing can differentially impact genomic, RNA, and protein integrity

  • Resolution Strategies:

    • Employ multiple detection methods for each level (genomic, transcript, protein)

    • Use isoform-specific primers and antibodies

    • Include appropriate controls for each method

    • Consider the biological relevance of each measurement (functional consequences)

  • Integrated Analysis Framework:

    • Start with genomic alterations (deletions, mutations)

    • Correlate with transcript expression patterns (full-length vs. isoforms)

    • Validate with protein expression and subcellular localization

    • Confirm functional impact through downstream target analysis

This comprehensive approach helps distinguish biological complexity from technical artifacts, providing more robust interpretation of IKZF1 status.

What in vivo models best represent human IKZF1-related pathologies for therapeutic testing?

Several in vivo models effectively represent human IKZF1-related pathologies:

  • Xenograft Models:

    • Patient-derived xenografts (PDX) from IKZF1-altered leukemias

    • Cell line xenografts with engineered IKZF1 alterations

    • Advantages: Human genetic context; reflects patient heterogeneity

    • Limitations: Immunodeficient host environment; limited immune interaction studies

  • Genetic Mouse Models:

    • Ikzf1 knockout/knockin models (global or conditional)

    • Models with specific mutations (e.g., R183C/H knockin)

    • Advantages: Intact immune system; developmental effects can be studied

    • Limitations: Species differences in IKZF1 function and regulation

  • Humanized Models:

    • Reconstitution of immunodeficient mice with human hematopoietic system

    • Introduction of IKZF1 alterations to human CD34+ cells

    • Advantages: Human hematopoietic development in vivo; immune interaction studies

    • Limitations: Incomplete human immune development; technical complexity

When selecting models for therapeutic testing, researchers should consider:

  • Match between model and specific IKZF1 alteration being studied

  • Relevant endpoints (survival, blast counts, organ infiltration)

  • Pharmacokinetic considerations for the therapeutic agent

  • Appropriate controls (isogenic cells with wild-type IKZF1)

The most robust approach combines multiple complementary models to validate therapeutic findings across different experimental systems.

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.