POU3F2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 working days after receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery information.
Synonyms
Brain 2 antibody; Brain specific homeobox/POU domain protein 2 antibody; Brain-2 antibody; Brain-specific homeobox/POU domain protein 2 antibody; BRN 2 antibody; Brn-2 antibody; BRN2 antibody; N Oct 3 antibody; N OCT 3 Gene antibody; N Oct3 antibody; Nervous system specific octamer binding transcription factor antibody; Nervous system-specific octamer-binding transcription factor N-Oct-3 antibody; OCT 7 antibody; Oct-7 antibody; OCT7 antibody; Octamer Binding Transcription Factor 7 antibody; Octamer-binding protein 7 antibody; Octamer-binding transcription factor 7 antibody; OTF 7 antibody; OTF-7 antibody; OTF7 antibody; PO3F2_HUMAN antibody; POU class 3 homeobox 2, antibody; POU domain class 3 transcription factor 2 antibody; POU domain, class 3, transcription factor 2 antibody; Pou3f2 antibody; POUF 3 antibody; POUF3 antibody; Protein Brn 2 antibody
Target Names
POU3F2
Uniprot No.

Target Background

Function
POU3F2 is a transcription factor that plays a crucial role in neuronal differentiation. It exhibits preferential binding to a recognition sequence composed of two distinct half-sites, ('GCAT') and ('TAAT'), separated by a non-conserved spacer region of 0, 2, or 3 nucleotides. When acting as a transcriptional activator, it cooperatively binds with SOX4, SOX11, or SOX12 to gene promoters. The combination of three transcription factors, ASCL1, POU3F2/BRN2, and MYT1L, is sufficient to reprogram fibroblasts and other somatic cells into induced neuronal (iN) cells in vitro. Operating downstream of ASCL1, POU3F2 accesses chromatin opened by ASCL1 and promotes the transcription of neuronal genes.
Gene References Into Functions
  1. Studies indicate that BRN2 is directly transcriptionally repressed by AR, essential for the expression of terminal neuroendocrine markers and aggressive growth of castration-resistant prostate cancer (CRPC), and highly expressed in human neuroendocrine prostate cancer and metastatic CRPC with low circulating PSA. PMID: 27784708
  2. These findings not only shed light on the molecular mechanism of the anticancer properties of capsaicin but also the transcription regulation of tNOX expression, potentially explaining the association of POU3F2 with tumorigenesis. PMID: 27271588
  3. Our research demonstrates that POU3F2 operates downstream of SIM1 and controls oxytocin expression in the hypothalamic neuroendocrine preoptic area. This finding aligns with the expression patterns of POU3F2 and related genes in the human brain. PMID: 26833329
  4. TTF1 expression in small-cell lung cancer is a cell lineage-specific phenomenon involving the developing neural cell-specific homeoprotein BRN2. PMID: 23358112
  5. Our study investigated the transcriptional regulation of CDH13 in melanoma. We found an inverse correlation between BRN2 and T-cadherin protein and transcript expression in melanoma. PMID: 23069940
  6. Our research links BRN2 as an activator and MITF as a repressor of the NOTCH pathway in melanoma cells. Loss of the BRN2-MITF axis in antisense-ablated cell lines decreased the melanoma sphere-forming capability and cell adhesion. PMID: 21358674
  7. We propose a model for the apparent non-overlapping expression levels of BRN2 and MITF in melanoma, mediated by miR-211 expression. PMID: 21435193
  8. POU3F2 may regulate the properties of melanoma cells via autocrine KIT ligand signaling. PMID: 20337985
  9. Our findings suggest that the high levels of Brn-2 expression observed in melanomas link BRAF signaling to increased proliferation. PMID: 15024080
  10. Rb induces a proliferative arrest and regulates Brn-2 expression in retinoblastoma cells. PMID: 17163992
  11. Brn-2 regulates invasiveness and directly represses Mitf expression. PMID: 18829533
  12. SOX9 and SOX10 but not BRN2 appear to be required for nestin expression in human melanoma. PMID: 18923447
  13. We report the expression and purification of full-length BRN2, a transcription factor implicated in melanoma growth. PMID: 18996486
Database Links

HGNC: 9215

OMIM: 600494

KEGG: hsa:5454

STRING: 9606.ENSP00000329170

UniGene: Hs.182505

Protein Families
POU transcription factor family, Class-3 subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Expressed specifically in the neuroectodermal cell lineage.

Q&A

What is POU3F2 and why is it significant in neuroscience research?

POU3F2 (POU domain, class 3, transcription factor 2), also known as BRN2 or N-Oct3, is a critical transcription factor predominantly expressed in the central nervous system. This 46.9 kDa protein plays essential roles in:

  • Neural development and patterning of the embryonic brain

  • Neuronal differentiation and cell fate determination

  • Regulation of cortical neural migration and neurogenesis

  • Establishment of neural cell lineages, particularly in the neocortex

The significance of POU3F2 extends to its ability to convert fibroblasts to functional neurons when combined with other transcription factors (ASCL1 and MYT1L), forming induced neuronal (iN) cells. This capacity makes POU3F2 particularly valuable for research into neurodegenerative diseases and neuronal development .

How do POU3F2 expression patterns change during neural development?

POU3F2 shows dynamic expression patterns throughout neural development:

  • Early embryonic stages: Upregulated in progenitor cells of the subventricular zone, intermediate zone, and outer layer of the neocortex

  • Mid-development: Contributes to neural formation and cell fate determination

  • Later stages: Regulates cortical neural migration and neurogenesis

Studies using knockout models have demonstrated that inactivation of the Pou3f2 gene in mice results in complete loss of development of specific neuronal lineages in paraventricular nuclei and supraoptic nuclei in the hypothalamus, highlighting its critical developmental role .

What is the molecular mechanism of POU3F2 function in neuronal differentiation?

POU3F2 functions through specific molecular mechanisms:

MechanismDescriptionDownstream Effects
DNA bindingBinds preferentially to recognition sequences with two distinct half-sites, 'GCAT' and 'TAAT', separated by nonconserved spacer regionsActivates target gene transcription
Target gene regulationRegulates genes like NTF3 (neurotrophin-3) and miR-320ePromotes neural development and differentiation
Regulatory networksFunctions as a key regulator in gene coexpression networksImpacts neuropsychiatric disorder susceptibility

In neural progenitor cells, POU3F2 knockdown studies have shown increased cell proliferation (EdU+/DAPI+ ratio) and decreased differentiation to neurons (reduced Tuj1+ and MAP2+ cells), demonstrating its role in balancing neural progenitor proliferation versus differentiation .

What criteria should be considered when selecting a POU3F2 antibody for specific applications?

When selecting a POU3F2 antibody, researchers should consider several critical parameters:

  • Target epitope region:

    • N-terminal antibodies (e.g., targeting Asn7-His59) typically recognize full-length N-Oct3 isoform

    • C-terminal antibodies may detect multiple isoforms

    • Middle region antibodies (e.g., targeting amino acids 199-226) detect the conserved functional domains

  • Validation for specific applications:

    • For Western blot: Confirm detection of appropriate molecular weight band (~47 kDa)

    • For ChIP applications: Verify chromatin immunoprecipitation efficiency with known target genes

    • For immunofluorescence: Ensure nuclear localization pattern in neural tissues

  • Species cross-reactivity:

    • Human/mouse/rat cross-reactive antibodies are valuable for comparative studies

    • The immunogen sequence used for antibody generation should be analyzed for cross-species conservation

  • Clonality considerations:

    • Monoclonal antibodies offer higher specificity and reproducibility between lots

    • Polyclonal antibodies may provide higher sensitivity but with potential batch variation

How should researchers validate POU3F2 antibodies for chromatin immunoprecipitation experiments?

Proper validation of POU3F2 antibodies for ChIP experiments requires a systematic approach:

  • Preliminary validation:

    • Western blot analysis to confirm antibody specificity

    • Immunofluorescence to verify nuclear localization pattern

  • ChIP optimization:

    • Sonication parameters: 40 cycles (30s on/30s off) using 20% power amplitude has been successfully employed

    • Antibody amount: 5 μg POU3F2 antibody per immunoprecipitation is recommended

    • Include appropriate IgG controls (naive IgG mixed with Dynabeads protein-G)

  • Validation of known targets:

    • PCR amplification of known POU3F2 binding sites (e.g., NTF3 promoter)

    • Quantitative PCR using primers flanking predicted binding sites

  • ChIP-seq considerations:

    • Cross-link cells with 1% formaldehyde

    • Sonicate chromatin to 200bp-1kb fragments

    • Sequence using high-throughput methods like Illumina sequencing

    • Analyze with appropriate peak-calling algorithms

Successful ChIP experiments have been reported using antibodies like sc-6029 (Santa Cruz Biotechnology), demonstrating POU3F2 binding to promoter regions of target genes such as NTF3 .

What are the optimal conditions for detecting POU3F2 by Western blot analysis?

Successful Western blot detection of POU3F2 requires careful optimization:

ParameterRecommended ConditionsNotes
Protein preparationCell lysis in RIPA buffer with protease inhibitorsCentrifuge at 12,000g for 15 min to remove debris
Sample loading20-50 μg total proteinLoad control samples (e.g., brain tissue lysate)
Gel percentage10% SDS-PAGEResolves the 47 kDa POU3F2 protein effectively
Transfer conditionsPVDF membraneProvides better protein retention than nitrocellulose
Blocking conditions5% non-fat milk in TBS-T1 hour at room temperature
Primary antibody1-2 μg/mLOvernight incubation at 4°C
Detection systemHRP-conjugated secondary antibody1 hour incubation followed by chemiluminescence imaging

Expected results should show a specific band at approximately 47 kDa, though some antibodies may detect bands at slightly higher molecular weights (up to 68 kDa) depending on post-translational modifications or isoform detected.

A specific positive control is recommended - A375, Bowes, or SK-Mel-28 human melanoma cell lines have been validated for POU3F2 expression .

How can researchers effectively design knockdown experiments to study POU3F2 function?

Designing effective POU3F2 knockdown experiments requires consideration of several factors:

  • Knockdown method selection:

    • siRNA: For transient knockdown (41-51% reduction in expression observed in published studies)

    • shRNA: For stable knockdown in long-term differentiation studies

    • CRISPR-Cas9: For complete gene knockout when appropriate

  • Experimental design considerations:

    • Include appropriate controls (scrambled siRNA/shRNA)

    • Validate knockdown efficiency by qPCR and Western blot

    • Monitor downstream effects on target genes (e.g., hsa-miR-320e, NTF3)

    • Assess functional outcomes (proliferation, differentiation)

  • Cell model selection:

    • NT2D1 cells: Human pluripotent embryonic carcinoma line suitable for neuronal differentiation studies

    • Neural progenitor cells (NPCs): Primary cells for developmental studies

    • SH-SY5Y: Neuroblastoma line used for mechanistic studies

  • Functional analysis:

    • Proliferation assay: EdU incorporation (EdU+/DAPI+ ratio)

    • Differentiation markers: Tuj1 (immature neurons), MAP2 (mature neurons)

    • Gene expression analysis: qPCR for target genes

POU3F2 knockdown has been shown to increase neural progenitor proliferation while inhibiting their differentiation into neurons, effects that can be partially rescued by recombinant NTF3 supplementation .

How can POU3F2 antibodies be utilized in studies of neuropsychiatric disorders?

POU3F2 antibodies offer valuable tools for investigating neuropsychiatric disorders:

  • Postmortem brain tissue analysis:

    • Immunohistochemistry to compare POU3F2 expression patterns between control and diseased brain tissue

    • Co-immunostaining with other markers to identify affected neural populations

    • Quantitative analysis of nuclear POU3F2 levels in specific brain regions

  • Regulatory network investigation:

    • ChIP-seq to identify genome-wide binding patterns in control vs. disease models

    • Integration with transcriptomic data to construct gene coexpression networks

    • Identification of dysregulated POU3F2 target genes

  • Genetic variant functional studies:

    • Analysis of how disease-associated variants affect POU3F2 binding

    • Reporter assays to test variant effects on target gene expression

    • CRISPR-edited cellular models expressing disease-associated variants

Research has identified POU3F2 as a key regulator in a psychosis-associated brain gene expression module enriched for rare coding variants in schizophrenia-associated genes. Studies show it regulates expression of genes in brains of schizophrenia and bipolar disorder patients, functioning as a core regulator of gene coexpression networks underlying these disorder risks .

What are the technical challenges in studying POU3F2 interactions with target genes?

Studying POU3F2 interactions with target genes presents several technical challenges:

  • Binding site complexity:

    • POU3F2 binds to sequences with two distinct half-sites ('GCAT' and 'TAAT')

    • Variable spacing between half-sites (0, 2, or 3 nucleotides) complicates prediction

    • Binding often occurs in conjunction with other transcription factors

  • Methodological limitations:

    • ChIP-seq resolution limitations in identifying precise binding sites

    • Potential for antibody cross-reactivity with related POU family members

    • Challenges in distinguishing direct vs. indirect gene regulation

  • Validation strategies:

    • Promoter deletion/mutation studies: Unidirectional deletion or mutation of binding sites affects promoter-driven luciferase activity

    • Bidirectional validation: Both knockdown and overexpression should produce opposite effects on target gene expression

    • Multiple experimental systems: Validate in different cell types and in vivo models

For example, the regulatory relationship between POU3F2 and NTF3 was confirmed through multiple approaches:

  • ChIP-seq identified POU3F2 binding to the NTF3 promoter

  • Promoter deletion/mutation studies showed decreased luciferase activity

  • POU3F2 knockdown downregulated NTF3 expression

  • Recombinant NTF3 rescued effects of POU3F2 knockdown

  • Immunostaining showed colocalization in developing mouse neurons

How should researchers address conflicting antibody validation results?

When faced with conflicting antibody validation results, researchers should implement a systematic troubleshooting approach:

  • Technical validation:

    • Test multiple antibody dilutions and incubation conditions

    • Include appropriate positive and negative controls

    • Compare antibodies targeting different epitopes of POU3F2

    • Perform parallel validation with orthogonal methods (e.g., tagged protein expression)

  • Statistical considerations:

    • Replicate experiments multiple times with different antibody lots

    • Quantify signal-to-noise ratios under different conditions

    • Perform statistical analysis to determine significance of observed differences

  • Resolution strategies:

    • For Western blot discrepancies: Confirm predicted molecular weight (46.9 kDa) versus observed band pattern

    • For immunostaining differences: Verify specificity with knockdown/knockout controls

    • For ChIP inconsistencies: Validate with known binding sites and consensus sequences

  • Documentation and reporting:

    • Document all validation results, even contradictory ones

    • Report antibody catalog numbers, lots, and detailed experimental conditions

    • Consider cell type and tissue-specific differences in POU3F2 expression patterns

What are the key considerations for interpreting POU3F2 expression patterns in neural development studies?

Interpreting POU3F2 expression patterns in neural development requires careful consideration:

  • Developmental timing:

    • POU3F2 expression is dynamically regulated during development

    • Expression peaks in progenitor cells during early neurogenesis

    • Changes in expression timing may indicate developmental abnormalities

  • Spatial distribution:

    • Normal expression is primarily in the subventricular zone, intermediate zone, and outer layers of developing neocortex

    • Compare expression between multiple brain regions and across developmental stages

    • Co-localization with stage-specific markers provides context for interpretation

  • Quantitative analysis:

    • Measure both percentage of POU3F2-positive cells and expression intensity

    • Compare against appropriate developmental stage-matched controls

    • Analyze in context of proliferation markers (EdU) and differentiation markers (Tuj1, MAP2)

  • Functional relevance assessment:

    • Correlate POU3F2 expression changes with phenotypic outcomes

    • Analyze downstream target gene expression (NTF3, miR-320e)

    • Consider compensatory mechanisms involving other POU-domain transcription factors

Studies have shown that POU3F2 knockdown increases neural progenitor proliferation and decreases neuronal differentiation, indicating its role in balancing proliferation versus differentiation during neural development .

How does POU3F2 function within the broader context of transcriptional networks governing neuronal differentiation?

POU3F2 operates within complex transcriptional networks:

Network ComponentInteraction with POU3F2Functional Outcome
ASCL1 (Mash1)Cooperative activityNeuronal reprogramming of fibroblasts to iN cells
MYT1LSynergistic actionConversion of somatic cells to functional neurons
NTF3Direct transcriptional regulationPromotion of neuronal survival and differentiation
miR-320eUpstream regulationModulation of downstream neuronal gene expression
PAX6, ZNF423, SOX9Upstream regulationCoordination of neural development pathways

POU3F2 has been identified as a hub in gene coexpression networks underlying neuropsychiatric disorders. Network analysis (NEO results) indicates POU3F2 acts as an upstream regulator of multiple transcription factors (PAX6, ZNF423, SOX9) and miRNAs (hsa-miR-320e), forming a hierarchical regulatory cascade during neuronal differentiation .

What methodological approaches can integrate POU3F2 antibody-based studies with systems neuroscience?

Integrating POU3F2 antibody studies with systems neuroscience requires multidisciplinary approaches:

  • Multi-omics integration:

    • Combine ChIP-seq data with RNA-seq to correlate binding with expression changes

    • Integrate proteomics data to identify POU3F2 interaction partners

    • Apply computational models like Network Edge Orienting (NEO) to infer causal relationships

  • Single-cell resolution techniques:

    • Single-cell ChIP-seq to identify cell-type-specific binding patterns

    • Single-cell RNA-seq combined with trajectory analysis to map POU3F2's role in cell fate decisions

    • Spatial transcriptomics to correlate POU3F2 activity with anatomical position

  • In vivo validation approaches:

    • Conditional knockout models with temporally and spatially restricted POU3F2 deletion

    • In utero electroporation for region-specific manipulation

    • CRISPR-based lineage tracing to follow cells with modified POU3F2 activity

  • Translational applications:

    • Patient-derived iPSC models for neuropsychiatric disorders

    • Drug screening targeting POU3F2 regulatory networks

    • Correlation of genetic variants with POU3F2 binding and function

Studies have successfully applied these approaches to identify POU3F2 as a key regulator in psychosis-associated brain gene expression modules, demonstrating its importance in schizophrenia and bipolar disorder risk. Computational models like Network Edge Orienting (NEO) have been particularly useful in inferring causal relationships between POU3F2 and its targets .

What are the optimal storage conditions for maintaining POU3F2 antibody activity?

Proper storage is crucial for maintaining antibody functionality:

  • Temperature recommendations:

    • Long-term storage: -20°C to -70°C (up to 12 months from receipt)

    • Medium-term storage: -20°C in small aliquots to prevent freeze-thaw cycles (up to 6 months)

    • Short-term storage: 2-8°C under sterile conditions (up to 1 month after reconstitution)

  • Aliquoting strategy:

    • Divide antibody into single-use aliquots immediately upon receipt

    • Use small volumes (10-50 μL) to minimize freeze-thaw cycles

    • Store in sterile, contaminant-free tubes with proper sealing

  • Buffer considerations:

    • Most commercial POU3F2 antibodies are supplied in PBS with 0.09% sodium azide

    • Avoid repeated freeze-thaw cycles which can cause protein denaturation

    • Consider adding carrier proteins (BSA) for dilute antibody solutions

  • Quality control measures:

    • Document date of receipt and thawing

    • Periodically test functionality on positive control samples

    • Include detailed handling records in experimental documentation

How should researchers validate aging POU3F2 antibodies to ensure continued specificity?

Validating aging antibodies requires systematic assessment:

  • Comparison testing:

    • Compare performance between fresh and aged antibody lots

    • Test multiple dilutions to assess sensitivity changes

    • Evaluate signal-to-noise ratio in standard applications

  • Specificity assessment:

    • Western blot: Verify correct molecular weight band (47 kDa) and absence of non-specific bands

    • Immunofluorescence: Confirm nuclear localization pattern in neural tissues

    • Include positive controls (e.g., A375, Bowes, or SK-Mel-28 human melanoma cell lines)

    • Use negative controls (tissues/cells with low POU3F2 expression)

  • Functional validation:

    • For ChIP applications: Verify enrichment of known target sequences (NTF3 promoter)

    • For immunoprecipitation: Confirm pull-down of POU3F2 protein

    • Include blocking peptide controls when available

  • Rejuvenation strategies:

    • Avoid further dilution of aging antibodies

    • Consider concentration using appropriate molecular weight cut-off filters

    • Document changes in optimal working concentration over time

How might recent advances in antibody technology enhance POU3F2 research?

Emerging antibody technologies offer new opportunities for POU3F2 research:

  • Recombinant antibody development:

    • Single-chain variable fragments (scFvs) for improved tissue penetration

    • Intrabodies for live-cell tracking of POU3F2

    • Nanobodies for super-resolution microscopy applications

  • Proximity labeling techniques:

    • TurboID or APEX2 fusion antibodies for identifying POU3F2 interaction partners

    • Split-BioID systems to study context-specific interactions

    • Spatially-resolved proteomic mapping of POU3F2 complexes

  • Multifunctional antibody reagents:

    • BiTE (Bispecific T-cell Engager) technology for targeted manipulation

    • Antibody-DNA conjugates for spatial genomics applications

    • Antibody-PROTAC conjugates for targeted protein degradation

  • In vivo applications:

    • Blood-brain barrier-penetrating antibody derivatives

    • Optogenetic antibody systems for temporal control

    • Antibody-based biosensors for real-time activity monitoring

What emerging research questions about POU3F2 function could be addressed using antibody-based approaches?

Several emerging research questions can be addressed using antibody-based approaches:

  • Post-translational modification landscape:

    • How do phosphorylation, SUMOylation, or other modifications affect POU3F2 function?

    • Which enzymes regulate these modifications?

    • How do modifications change during development or in disease states?

  • Dynamic nuclear localization:

    • What regulates POU3F2 nuclear import/export?

    • How does localization change during cell cycle or differentiation?

    • What protein interactions govern subcellular distribution?

  • Chromatin remodeling activity:

    • How does POU3F2 interact with chromatin remodeling complexes?

    • What is the temporal sequence of transcription factor binding and chromatin changes?

    • How do pioneer factor activities contribute to neuronal fate specification?

  • Single-cell heterogeneity:

    • How variable is POU3F2 expression within seemingly homogeneous neural populations?

    • Does POU3F2 expression level correlate with cell fate choices?

    • Can antibody-based FACS sorting identify functionally distinct neural progenitor subpopulations?

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.