FOXA1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Generally, we can ship your orders within 1-3 business days of receipt. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
forkhead box A1 antibody; Forkhead box protein A1 antibody; FOX A1 antibody; FOXA1 antibody; FOXA1_HUMAN antibody; hepatocyte nuclear factor 3 alpha antibody; Hepatocyte nuclear factor 3-alpha antibody; HNF 3A antibody; HNF-3-alpha antibody; HNF-3A antibody; HNF3A antibody; MGC33105 antibody; TCF 3A antibody; TCF-3A antibody; TCF3A antibody; Transcription factor 3A antibody
Target Names
Uniprot No.

Target Background

Function
FOXA1 is a transcription factor that plays a crucial role in embryonic development, establishing tissue-specific gene expression, and regulating gene expression in differentiated tissues. It is considered a 'pioneer' factor, capable of opening compacted chromatin for other proteins by interacting with nucleosomal core histones and replacing linker histones at target enhancer and/or promoter sites. FOXA1 binds to DNA with the consensus sequence 5'-[AC]A[AT]T[AG]TT[GT][AG][CT]T[CT]-3'. It is believed to play a role in translating epigenetic signatures into cell type-specific enhancer-driven transcriptional programs. Its differential recruitment to chromatin is dependent on the distribution of histone H3 methylated at 'Lys-5' (H3K4me2) in estrogen-regulated genes. FOXA1 is involved in the development of multiple endoderm-derived organ systems such as liver, pancreas, lung, and prostate; FOXA1 and FOXA2 seem to have at least partially redundant roles. It modulates the transcriptional activity of nuclear hormone receptors. FOXA1 is involved in ESR1-mediated transcription; it is required for ESR1 binding to the NKX2-1 promoter in breast cancer cells; it binds to the RPRM promoter and is required for the estrogen-induced repression of RPRM. FOXA1 is also involved in regulating apoptosis by inhibiting the expression of BCL2 and in cell cycle regulation by activating expression of CDKN1B, alone or in conjunction with BRCA1. FOXA1 was initially described as a transcription activator for several liver genes, including AFP, albumin, tyrosine aminotransferase, PEPCK, etc. It interacts with the cis-acting regulatory regions of these genes. It is also involved in glucose homeostasis.
Gene References Into Functions
  1. High FOXA1 expression is associated with the pathogenesis of gastric cancer. PMID: 30066917
  2. FoxA1 might be related to the development and differentiation of secretory coil-like structures, as well as the secretory function of the 3D reconstructed eccrine sweat glands. PMID: 29909922
  3. TUG1 promoted osteosarcoma cell proliferation and suppressed apoptosis by regulating the miR-212-3p/FOXA1 axis. PMID: 29793327
  4. miR-760 should be considered as a tumor suppressor since it negatively regulates the oncogene protein FOXA1 and regulated TRAIL sensitivity in Non-small cell lung cancer cells. PMID: 29665655
  5. As a tumor suppressor, FOXA1 targets PIK3R1 directly to inhibit PI3K/Akt signaling pathway, thus exerting a negative regulatory effect on proliferation, migration, and invasion of HCC in male patients. PMID: 29208003
  6. miR212 suppresses renal cell carcinoma (RCC) proliferation and invasion by modulating FOXA1, suggesting that miR212 may have potential as a therapeutic target in RCC. PMID: 29115609
  7. the results of the present study suggested that FOXA1 is a potential oncogene in NSCLC PMID: 29115441
  8. During the ensuing weeks, the PAX2/FOXA1 boundary progressively extended cranially such that by 21 weeks the entire vaginal epithelium was FOXA1-reactive and PAX2-negative. This observation supports Bulmer's proposal that human vaginal epithelium derives solely from urogenital sinus epithelium PMID: 28918284
  9. The transcription factor FOXA1 directly bound to the PLOD2 promoter, and turned on PLOD2 transcription. In summary, our findings revealed a regulatory mechanism of NSCLC metastasis through EGFR-PI3K/AKT-FOXA1-PLOD2 pathway. PMID: 29072684
  10. maintenance of the cancer cell state is dependent on recruitment of Mediator and Cohesin through FOXA and master transcription factors PMID: 27739523
  11. overexpression of GATA3 and FOXA1 cooperate with PPAR activation to drive transdifferentiation of a basal bladder cancer cells to a luminial phenotype. PMID: 27924948
  12. Results found FOXA1 to be hypermethylated in breast tumors from African American (AA) versus European American (EA) women with ER- cancer, and methylation levels showed strong inverse relationships with both mRNA and protein levels. A significant positive association was identified between parity and FOXA1 methylation in tumors from AA women who did not breastfeed. PMID: 28756535
  13. High expression of FOXA1 is associated with breast tumor. PMID: 28534958
  14. High FOXA1 expression was significantly associated with presence of lymph node metastasis (LNM), low tumour-infiltrating lymphocytes (TILs), and submucosal invasion. PMID: 28843920
  15. Results provide evidence that the atypical expression of FOXA1 transcriptional factor is an important player in psoriasis, as it inhibits maturation of naive T cells into a Treg subpopulation, therefore contributing to the development of psoriatic skin lesions. PMID: 28849568
  16. Authors demonstrated that ER(alpha), GATA3 and FOXA1 form a transcriptional complex with Ell3 to regulate IL-20 expression in ER(+) breast cancer cells. FOXA1 represses IL-20 expression, whereas GATA3 and ER(alpha) activate it. PMID: 28514748
  17. the findings show that squamous and micropapillary bladder cancers have different expression patterns of CK14 and FOXA1 and suggest that they arise from distinct precursors. PMID: 28721490
  18. rs4442975 may not confer a risk of breast cancer occurrence or progression in the Chinese Han population. PMID: 27835577
  19. As a transcriptionally regulated program, urothelial differentiation operates as a heterarchy, wherein GATA3 is able to co-operate with FOXA1 to drive expression of luminal marker genes, but that P63 has potential to transrepress expression of the same genes. PMID: 28282036
  20. FOXA1 overexpression mediates endocrine resistance by altering the ER transcriptome and IL-8 expression in ER-positive breast cancer. PMID: 27791031
  21. Findings suggested FOXA1 may act as an anti-oncogene in gastric cancer (GC) cells. Low-level expression of FOXA1 protein was confirmed in GC tissues and cell lines. FOXA1 over-expression could significantly affect cell proliferation, apoptosis and tumor invasion of GC cells, which may be resulted by reversing EMT. PMID: 29129808
  22. High expression of FOXA1 is an independent prognostic parameter in ERG negative prostate cancer PMID: 29029032
  23. A molecular mechanism by which Estradiol antagonizes GR-dependent induction of specific genes by preventing the recruitment of the pioneer factors FOXA1 and FOXA2 in a physiologically relevant model. PMID: 28938408
  24. Results show that c-Abl phosphorylates FoxA1 at multiple sites. Tyr429 and Tyr464 were identified as the major phosphorylation sites in the FoxA1 C-terminal region. This c-Abl-mediated phosphorylation of FoxA1 promotes the activation of estrogen signaling by inducing its binding to histones. PMID: 27883218
  25. MLL3 binding was dependent on FOXA1, indicating that FOXA1 recruits MLL3 to chromatin. MLL3 silencing decreased H3K4me1 at enhancer elements but had no appreciable impact on H3K4me3 at enhancer elements. We propose a mechanism whereby the pioneer factor FOXA1 recruits the chromatin modifier MLL3 to facilitate the deposition of H3K4me1 histone marks, subsequently demarcating active enhancer elements PMID: 27926873
  26. FoxA1 discriminates between medullary thyroid carcinoma and tumors of follicular derivation with sensitivity and specificity greater than calcitonin and carcinoembryonic antigen. PMID: 28546130
  27. FOXA1 loss may play a significant role in enabling prostate cancer progression to neuroendocrine prostate cancer, whereas IL-8 and MAPK/ERK pathways may be promising targets for therapeutic intervention. PMID: 28319070
  28. Low FOXA1 expression is associated with breast cancer invasion and metastasis. PMID: 27524420
  29. the distinct mechanisms by which GATA2 and FOXA1 regulate AR cistrome and suggest that FOXA1 acts upstream of GATA2 and AR in determining hormone-dependent gene expression in prostate cancer. PMID: 26751772
  30. Study implicates enhancer reprogramming, FOXA1 upregulation, and a retrograde developmental transition in pancreatic ductal adenocarcinoma metastasis. PMID: 28757253
  31. Studies indicate that microRNA miR-212 exerts its inhibitory effect on hepatocellular carcinoma (HCC) by inhibiting forkhead transcription factor FOXA1 expression. PMID: 27999212
  32. FOXA1 is expressed by basal cells of squamous epithelium, pre-invasion lesions of the uterine cervix and the head/neck and almost half invasive cervical and head/neck carcinomas, supporting its possible implication in HPV associated carcinomas. PMID: 28209524
  33. CREB1/FoxA1 signaling is a targetable driver of prostate cancer progression and serves as a biomarker of poor clinical outcomes. PMID: 26743006
  34. miR-93 may promote the process of epithelial-mesenchymal transition in endometrial carcinoma cells by targeting FOXA1. PMID: 27829043
  35. conclusion, our results demonstrated that miR24 inhibits breast cancer cells invasion by targeting OGT and reducing FOXA1 stability. These results also indicated that OGT might be a potential target for the diagnosis and therapy of breast cancer metastasis. PMID: 28455227
  36. FOXA1 is not only able to recognize but also remodel the epigenetic signatures at lineage-specific enhancers, which is mediated, at least in part, by a feed-forward regulatory loop between FOXA1 and TET1. PMID: 27257062
  37. Based on these results, we suggest that FOXA1 plays a catalytic role in ovarian cancer pathogenesis and development by affecting the expression of the above-mentioned proteins. PMID: 28488543
  38. these results suggest that FOXA1 suppresses expression of IL6 through inhibition of NF-kappaB recruitment to the IL6 promoter in an ERalpha-independent manner and that reduction in FOXA1 expression induces IL6 expression and contributes to cancer stem cell-like properties in TAM-R cells. PMID: 28270510
  39. These results suggest a positional-nucleosome-oriented accessing model for Pioneer factors seeking target motifs, in which FOXA1 can examine each underlying DNA nucleotide. PMID: 27458208
  40. Down-regulation of TRPS1 by miR-373, acting as a transcriptional activator, promotes epithelial-mesenchymal transition (EMT) and metastasis by repressing FOXA1 transcription, expanding upon its previously reported role as a transcription repressor. PMID: 26969828
  41. FOXA1 is found in ovarian mucinous and Brenner tumours PMID: 28238418
  42. the results of the present study indicated that FOXA1 may be considered a potential prognostic marker, and may promote tumor growth of CRC by upregulating YAP expression. PMID: 27484093
  43. meta-analysis showed that high expression of FOXA1 in breast cancer patients was a good prognostic indicator for survival outcome. PMID: 27212698
  44. FOXA1 promotes cell senescence in EC by interaction with p16INK4a. PMID: 27349269
  45. There is significantly increased esophageal squamous cell carcinoma risk associated with the FOXA1 rs7144658 T > C polymorphism among male patients. PMID: 27050876
  46. Meta-analysis results clearly point to an important role of FOXA1 and FOXA2 gene regulatory networks in the etiology of Alzheimer's disease PMID: 26890743
  47. show that ER and GR both have the ability to alter the genomic distribution of the FoxA1 pioneer factor. Single-molecule tracking experiments reveal a highly dynamic interaction of FoxA1 with chromatin in vivo; FoxA1 factor is not associated with footprints at its binding sites throughout the genome; findings support a model wherein interactions between transcription factors and pioneer factors are highly dynamic. PMID: 27062924
  48. findings suggest a key role for GalNAc-T4 in the estrogen pathway through FOXA1 glycosylation PMID: 26541755
  49. The ratio of FoxA1 to FoxA2 in lung adenocarcinoma is regulated by LncRNA HOTAIR and chromatin remodeling factor LSH PMID: 26658322
  50. Loss of Interdependent Binding by the FoxO1 and FoxA1/A2 Forkhead Transcription Factors Culminates in Perturbation of Active Chromatin Marks and Binding of Transcriptional Regulators at Insulin-sensitive Genes. PMID: 26929406

Show More

Hide All

Database Links

HGNC: 5021

OMIM: 602294

KEGG: hsa:3169

STRING: 9606.ENSP00000250448

UniGene: Hs.163484

Subcellular Location
Nucleus.
Tissue Specificity
Highly expressed in prostate and ESR1-positive breast tumors. Overexpressed in esophageal and lung adenocarcinomas.

Q&A

What is FOXA1 and why is it an important research target?

FOXA1 (Forkhead Box A1) is a transcription factor belonging to the FOX family, originally identified as hepatocyte nuclear factor 3-alpha (HNF-3-alpha). It functions as a pioneering factor that can open compact chromatin structures to facilitate binding of other proteins, playing crucial roles in:

  • Embryonic development and tissue differentiation

  • Establishment of tissue-specific gene expression

  • Regulation of gene expression in differentiated tissues

  • Cell cycle regulation through activation of CDKN1B

  • Translating epigenetic signatures into enhancer-driven lineage-specific transcriptional programs

The human FOXA1 protein consists of 472 amino acid residues with a molecular mass of 49.1 kDa and is primarily localized in the nucleus. It is highly expressed in prostate and ESR1-positive breast tumors, making it a significant target in cancer research .

How do I select the appropriate FOXA1 antibody for my specific experimental application?

Selection of the optimal FOXA1 antibody depends on your experimental application, target species, and cellular compartment of interest. Consider these methodological criteria:

Application compatibility:

ApplicationRecommended Antibody TypeNotes
Western BlotMonoclonal or polyclonalUse 1:1000-1:4000 dilution; for best results, avoid freeze-thaw cycles of lysates
IHC-PMonoclonal preferredRequires antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0
IF/ICCEither typeUse 1:50-1:500 dilution
ChIP/ChIP-seqChIP-grade polyclonalSpecially validated for chromatin studies
Flow CytometryFluorophore-conjugatedTrue-Nuclear™ Transcription Factor Buffer Set improves staining

Species reactivity: Most commercial antibodies are validated for human FOXA1, but some also react with mouse, rat, and other species. Check cross-reactivity if working with non-human models .

Epitope location: For studying protein-protein interactions or DNA binding, choose antibodies targeting regions outside the forkhead/winged helix domain to avoid interference with functional studies .

Always perform validation experiments in your specific model system before proceeding with full-scale experiments .

What are the optimal protocols for detecting nuclear FOXA1 expression by immunofluorescence?

Detecting nuclear FOXA1 by immunofluorescence requires careful attention to fixation, permeabilization, and imaging parameters:

Optimized protocol for robust nuclear FOXA1 detection:

  • Cell preparation:

    • Culture cells on glass coverslips or chamber slides

    • For MCF-7 or similar adherent cell lines, grow to 70-80% confluence

  • Fixation and permeabilization:

    • Fix cells with 1% paraformaldehyde (PFA) for 10 minutes at room temperature

    • For improved nuclear antigen access, use True-Nuclear™ Transcription Factor Buffer Set or similar nuclear transcription factor staining buffer systems

    • Alternative method: fix with 4% PFA followed by permeabilization with 0.1% Triton X-100 for 5-10 minutes

  • Blocking and antibody incubation:

    • Block with 5% BSA in PBS for 1 hour

    • Incubate with primary anti-FOXA1 antibody (1:50-1:500 dilution) for 3 hours at room temperature or overnight at 4°C

    • For visualization, use fluorophore-conjugated secondary antibodies or directly conjugated primary antibodies (e.g., Alexa Fluor 647 anti-FOXA1)

  • Nuclear counterstaining:

    • Counterstain with DAPI to clearly delineate nuclei

    • Mount using anti-fade mounting medium

  • Imaging considerations:

    • Use confocal microscopy for precise nuclear localization

    • Adjust exposure to avoid saturating signal from highly expressing cells

    • Include appropriate negative controls (isotype control or FOXA1-negative cell line such as HEL cells)

This protocol has been validated for detecting FOXA1 in MCF-7 breast cancer cells, showing specific nuclear localization .

How should FOXA1 antibodies be validated for specificity in ChIP and ChIP-seq applications?

Validation of FOXA1 antibodies for chromatin immunoprecipitation applications requires comprehensive controls to ensure specificity and reproducibility:

Multifaceted validation approach:

  • Western blot pre-validation:

    • Confirm single band detection at approximately 49-50 kDa

    • Use positive control cell lines known to express FOXA1 (MCF-7, HepG2, LNCaP)

    • Include negative control cell lines that don't express FOXA1

  • Critical ChIP-specific controls:

    • Input control: Reserve 5-10% of chromatin before immunoprecipitation

    • Negative control regions: Select genomic regions known not to bind FOXA1

    • Isotype control antibody: Use matched isotype IgG to assess non-specific binding

    • FOXA1 knockdown/knockout validation: Perform ChIP in FOXA1-depleted cells to confirm signal reduction

  • Positive control genomic targets:

    • Target known FOXA1 binding sites:

      • Promoter regions of albumin and transthyretin genes

      • ESR1-regulated genes in breast cancer cells

      • AR-regulated genes in prostate cancer cells

  • Cross-validation approaches:

    • Compare results with an alternative antibody recognizing a different FOXA1 epitope

    • Validate key peaks by ChIP-qPCR prior to sequencing

    • For ChIP-seq, confirm enrichment of the FOXA1 consensus motif (A(A/T)TRTT(G/T)RYTY) in peak regions

  • Technical validation:

    • Optimize sonication conditions to yield 200-500 bp fragments

    • Titrate antibody concentration (typically 2-5 μg per ChIP reaction)

    • Examine enrichment of histone H3K4me2 at FOXA1 binding sites as this modification is associated with FOXA1 recruitment

By implementing these validation steps, researchers can ensure the specificity and reliability of FOXA1 antibodies in chromatin immunoprecipitation studies .

How can FOXA1 antibodies be utilized to study its pioneer factor activity in different cellular contexts?

FOXA1's pioneer factor activity can be investigated using specialized antibody-based approaches that reveal chromatin access, co-factor recruitment, and lineage-specific functions:

Advanced techniques for pioneer factor analysis:

  • Sequential ChIP (ChIP-reChIP) methodology:

    • First ChIP: Immunoprecipitate with FOXA1 antibody

    • Second ChIP: Use antibodies against collaborating factors (ESR1, AR, or STAT1)

    • This approach reveals simultaneous co-occupancy of FOXA1 with other transcription factors

    • Implementation: Use stringent washing between ChIPs and validate recovery efficiency

  • FAIRE-seq with FOXA1 ChIP correlation:

    • Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE) identifies open chromatin regions

    • Integrate FAIRE-seq data with FOXA1 ChIP-seq

    • High FAIRE signal at FOXA1 binding sites indicates active chromatin opening

    • Analysis shows that high FAIRE FOXA1-binding sites are more likely to recruit other factors (e.g., AR in prostate cancer)

  • Histone modification landscapes at FOXA1 binding sites:

    • Perform parallel ChIP-seq for:

      • FOXA1

      • H3K4me2 (prerequisite for FOXA1 binding)

      • H3K27ac (active enhancers)

    • Analysis reveals how FOXA1 translates epigenetic signatures into lineage-specific enhancers

    • Finding: H3K4 dimethylation occurs proximal to FOXA1-binding sites and can precede FOXA1 binding

  • Inducible FOXA1 systems for temporal studies:

    • Generate cell lines with inducible FOXA1 expression

    • Perform time-course experiments with ChIP-seq to track:

      • Initial FOXA1 binding

      • Subsequent chromatin opening

      • Recruitment of secondary factors

    • Correlate with DNA methylation changes, as FOXA1 binding precedes loss of cytosine methylation during differentiation

  • Cell-type comparative analyses:

    • Compare FOXA1 binding patterns across different cell types (e.g., breast vs. prostate cancer cells)

    • Finding: Differential FOXA1 recruitment to chromatin occurs predominantly at distant enhancers rather than proximal promoters

    • This differential recruitment leads to cell-type specific changes in chromatin structure and collaboration with lineage-specific transcription factors

These methodologies reveal that FOXA1's pioneer factor activity is context-dependent and influenced by pre-existing epigenetic marks, particularly H3K4 dimethylation .

What is the significance of FOXA1's interaction with nuclear hormone receptors, and how can these interactions be studied using antibody-based techniques?

FOXA1's interactions with nuclear hormone receptors (particularly estrogen receptor (ER) and androgen receptor (AR)) are critical in hormone-responsive cancers and can be investigated using sophisticated antibody-based methods:

FOXA1-nuclear receptor interaction significance:

FOXA1 modulates ER and AR function in breast and prostate cancers by:

  • Acting as a pioneer factor to facilitate receptor binding to chromatin

  • Forming physical complexes with receptors

  • Directing receptors to specific genomic locations

  • Maintaining specific hormonal transcriptional programs

Advanced antibody-based approaches to study these interactions:

These methods reveal that FOXA1 is essential for maintaining tissue-specific hormone-responsive transcriptional programs, with implications for understanding and treating hormone-dependent cancers .

What factors can influence FOXA1 antibody performance in Western blot applications, and how can inconsistent results be addressed?

Western blot detection of FOXA1 can present several challenges that require careful optimization:

Common issues and troubleshooting approaches:

  • Inconsistent band detection:

    • Issue: Multiple bands or band shifts from expected 49-50 kDa

    • Solutions:

      • Avoid freeze-thaw cycles of lysates, which can significantly impact FOXA1 detection

      • Use fresh lysates whenever possible

      • Include phosphatase inhibitors as FOXA1 undergoes post-translational modifications

      • Run gradient gels (4-12%) to improve resolution

  • Weak signal strength:

    • Issue: Low signal despite confirmed FOXA1 expression

    • Solutions:

      • Optimize nuclear extraction protocols (FOXA1 is exclusively nuclear)

      • Use specialized nuclear extraction buffers with high salt concentration

      • Increase lysate concentration to 30 μg for nuclear extracts

      • Extended transfer times (1-2 hours) for efficient transfer of higher molecular weight proteins

  • High background issues:

    • Issue: Non-specific binding creating interpretation difficulties

    • Solutions:

      • Extended blocking (overnight at 4°C)

      • Use 5% BSA instead of milk for blocking

      • Increase washing duration and detergent concentration

      • Optimize primary antibody dilution (test range from 1:1000-1:4000)

  • Cell line considerations:

    • Issue: Inconsistent detection across cell lines

    • Solutions:

      • Include validated positive controls: MCF-7, HepG2, MDA-MB-468, T-47D, or LNCaP cells

      • Use HEL cells as negative control

      • For prostate cancer studies, compare cytoplasmic vs. nuclear extracts as FOXA1 should be enriched in nuclear fraction

  • Antibody selection guidance:

    • Different antibodies may perform differently based on:

      • Epitope location (N-terminal vs. C-terminal vs. internal regions)

      • Monoclonal vs. polyclonal properties

      • For confident results, validate findings with multiple antibodies recognizing different epitopes

Validation experiment for FOXA1 Western blot:
Researchers should run a validation panel with:

  • Nuclear extracts from positive control cell lines

  • Whole cell lysates from the same cells

  • Negative control cell line

  • Molecular weight marker

  • Loading control targeting nuclear protein (e.g., Lamin B)

This approach allows proper evaluation of antibody specificity and performance before proceeding with experimental samples .

How should researchers address contradictory FOXA1 immunohistochemistry staining patterns in different tissue samples?

Contradictory FOXA1 immunohistochemistry results can arise from technical, biological, and interpretive factors that require systematic evaluation:

Sources of discrepancy and resolution approaches:

  • Pre-analytical variables:

    • Fixation impact: Formalin oversensitivity can mask FOXA1 epitopes

      • Solution: Standardize fixation time (24 hours optimal)

      • Different fixatives may yield different results; document fixative used

    • Tissue processing: Prolonged processing can denature nuclear antigens

      • Maintain consistent processing protocols across samples

    • Storage effects: Antigen degradation in stored slides

      • Use freshly cut sections when possible or analyze all samples simultaneously

  • Antigen retrieval optimization:

    • FOXA1 detection is highly dependent on proper retrieval

    • Critical finding: Use TE buffer pH 9.0 for optimal results

    • Alternative: Citrate buffer pH 6.0 may work but with potentially lower sensitivity

    • Heat-induced epitope retrieval (pressure cooker method) often yields superior results compared to microwave methods

  • Antibody-specific considerations:

    • Different clones recognize different epitopes:

      • Some antibodies detect only specific FOXA1 isoforms

      • Clone-dependent sensitivity to post-translational modifications

    • Validation strategy: Test multiple antibodies on tissue microarrays containing known positive/negative controls

  • Tissue-specific expression patterns:

    • Biological variation: FOXA1 expression varies significantly between tissues

      • High expression: Prostate and ESR1-positive breast tumors

      • Variable expression: Endoderm-derived tissues

    • Context-dependent expression: Consider tissue microenvironment

    • Cellular heterogeneity: Expression may vary within different cell populations in the same tissue

  • Quantification and interpretation standardization:

    • Implement standardized scoring systems:

      • H-score method (0-300 scale combining intensity and percentage positive)

      • Allred score (0-8 scale)

    • Use digital pathology systems for objective quantification

    • Consider both intensity and subcellular localization (exclusively nuclear for FOXA1)

  • Validation experiment:

    • Multi-tiered approach:

      • Include tissue with known high FOXA1 expression (breast cancer)

      • Include negative control tissue

      • Process paired samples with and without primary antibody

      • If applicable, include FOXA1 knockdown/knockout tissue samples

      • Compare staining pattern with mRNA expression data when available

By implementing these systematic approaches, researchers can resolve contradictory staining patterns and ensure reliable, reproducible FOXA1 immunohistochemistry results across different tissue types and experimental conditions .

How can FOXA1 antibodies be used to investigate its role in immune regulation and cancer immunotherapy?

Recent discoveries highlight FOXA1's unexpected role in immune regulation, particularly in modulating antitumor immunity. Antibody-based approaches offer powerful tools to explore this emerging research area:

Innovative methodologies for immune regulation studies:

  • FOXA1-immune checkpoint axis investigation:

    • Key finding: FOXA1 prevents tumor immune evasion by inhibiting IFN-γ induced PD-L1 expression in nasopharyngeal carcinoma cells

    • Experimental approach:

      • Multiplex immunofluorescence with FOXA1, PD-L1, and STAT1 antibodies

      • Flow cytometry to quantify PD-L1 expression in FOXA1-manipulated tumor cells

      • ChIP assays to detect FOXA1 binding at PD-L1 promoter regions

  • Molecular mechanism exploration:

    • FOXA1 interacts with STAT1 to inhibit IRF1 expression and binding to PD-L1 promoter upon IFN-γ treatment

    • Techniques:

      • Co-immunoprecipitation with FOXA1 and STAT1 antibodies

      • Sequential ChIP to identify genomic loci co-occupied by these factors

      • Proximity ligation assay to visualize protein interactions in situ

  • Tumor-immune cell interaction studies:

    • Experimental design:

      • Co-culture FOXA1-manipulated tumor cells with activated CD8+ T cells

      • Analyze T cell apoptosis by flow cytometry

      • Assess cytotoxic effector molecule expression

      • Finding: Co-culture with FOXA1-silenced nasopharyngeal carcinoma cells promotes apoptosis of activated tumor-specific CD8+ T cells

  • In vivo immune modulation:

    • Sophisticated animal models:

      • Xenograft models with FOXA1-overexpressing tumors

      • Adoptive T cell therapy in immunodeficient mice

      • Analysis of immune infiltration in tumor microenvironment

      • FOXA1 overexpression increases sensitivity to adoptive T cell therapy in mouse models

  • Integrating with immunotherapy biomarker research:

    • Clinical correlation approaches:

      • Multiplex IHC on patient samples to correlate FOXA1 expression with:

        • Immune cell infiltration patterns

        • PD-L1 expression

        • Response to immune checkpoint inhibitors

      • Creation of spatial maps of FOXA1, immune markers, and checkpoint molecules

These approaches leverage antibody-based technologies to elucidate FOXA1's unexpected role in regulating tumor-immune interactions, potentially identifying new biomarkers and therapeutic targets for cancer immunotherapy .

How are FOXA1 antibodies being used to understand epigenetic reprogramming in developmental processes and disease states?

FOXA1's pioneering activity in chromatin remodeling makes it a critical factor in epigenetic reprogramming. Advanced antibody-based approaches provide insights into these complex processes:

Cutting-edge methodologies for epigenetic studies:

  • Temporal mapping of epigenetic landscapes:

    • Sequential ChIP-seq approach:

      • Track FOXA1 binding during cellular differentiation or disease progression

      • Correlate with dynamic changes in histone modifications (H3K4me2, H3K27ac)

      • Integrate with DNA methylation analysis

      • Discovery: FOXA1 binding precedes loss of cytosine methylation and acquisition of H3K4me2 during cell differentiation

  • Single-cell technologies:

    • scATAC-seq with FOXA1 ChIP correlation:

      • Map chromatin accessibility at single-cell resolution

      • Identify cell populations with FOXA1-dependent chromatin states

      • Correlate with FOXA1 binding patterns from bulk ChIP-seq

      • Identify heterogeneous cellular responses to FOXA1-mediated epigenetic changes

  • CUT&RUN and CUT&Tag advancements:

    • Advantages over traditional ChIP:

      • Requires fewer cells (1,000-50,000 vs. millions)

      • Better signal-to-noise ratio

      • Compatible with flash-frozen tissues

    • Application strategy:

      • Map FOXA1 binding sites with higher resolution

      • Combine with histone modification mapping

      • Implement in rare cell populations or limited clinical samples

  • Developmental context studies:

    • Pancreatic development model:

      • FOXA1 and FOXA2 cooperatively control pancreatic acinar and islet morphogenesis

      • The presence of at least one wild-type allele of FOXA2 can compensate for complete loss of FOXA1

      • Experimental approach:

        • Combined IHC/IF for FOXA1, FOXA2, and lineage markers

        • ChIP-seq to compare genomic occupancy in wild-type vs. mutant tissues

        • Integration with transcriptomic data to identify compensatory mechanisms

  • Long-range chromatin interaction analysis:

    • HiChIP with FOXA1 antibodies:

      • Maps 3D genome organization at FOXA1 binding sites

      • Identifies long-range enhancer-promoter interactions

      • Reveals how FOXA1 shapes higher-order chromatin structure

    • Integration with gene expression:

      • Correlate FOXA1-mediated chromatin loops with gene expression changes

      • Identify genes regulated through FOXA1-dependent enhancers

These sophisticated approaches reveal FOXA1's central role in translating epigenetic signatures into enhancer-driven lineage-specific transcriptional programs during development and disease, with implications for understanding cellular plasticity, differentiation, and pathological states .

What techniques combine antibody detection with genomic analysis to correlate FOXA1 mutations with protein expression patterns?

Integrating antibody-based detection with genomic analysis provides crucial insights into how FOXA1 mutations affect protein expression, localization, and function:

Integrated genomic-proteomic approaches:

  • Mutation-specific antibody development:

    • Generate antibodies against common FOXA1 mutations

    • Focus on recurrent mutations in the C-terminal region and forkhead domain

    • Apply in IHC to differentiate wild-type from mutant FOXA1 expression patterns

    • Validate specificity using cell lines with known FOXA1 mutation status

  • Laser capture microdissection with combined analysis:

    • Workflow:

      • Perform IHC on tissue sections to identify FOXA1-positive regions

      • Use laser capture to isolate these specific regions

      • Split the sample for:

        • Targeted DNA sequencing of FOXA1

        • Protein extraction and analysis

      • Correlate mutation status with protein expression levels

  • Digital spatial profiling:

    • Use multiplex antibody panels including:

      • FOXA1

      • Downstream targets

      • Collaborating factors (ER, AR)

      • Epigenetic marks

    • Integrate with in situ hybridization for mutational analysis

    • Create spatial maps correlating mutations with protein expression patterns

  • CRISPR-engineered isogenic cell line panels:

    • Generate cell lines with common FOXA1 mutations

    • Perform comprehensive antibody-based analyses:

      • Western blot for expression levels

      • ChIP-seq for genomic binding patterns

      • Co-IP for altered protein interactions

    • Correlate findings with isogenic wild-type cells to identify mutation-specific effects

  • Patient-derived organoids with integrated analysis:

    • Establish organoid cultures from patient samples

    • Sequence for FOXA1 mutations

    • Perform immunofluorescence and ChIP-seq

    • Create isogenic corrected lines to directly assess mutation impact

    • This approach maintains tissue architecture context while enabling molecular analysis

These integrated approaches provide mechanistic insights into how FOXA1 mutations alter protein function, with implications for understanding disease progression and developing targeted therapies based on specific mutation profiles.

How might new antibody engineering technologies enhance FOXA1 research in complex tissue environments?

Emerging antibody technologies are poised to revolutionize FOXA1 research, particularly in complex in vivo settings and heterogeneous tissues:

Next-generation antibody technologies:

  • Nanobodies and single-domain antibodies:

    • Advantages for FOXA1 research:

      • Smaller size (15 kDa vs. 150 kDa) enables better tissue penetration

      • Access to hidden epitopes within protein complexes

      • Improved access to nuclear antigens

    • Applications:

      • Super-resolution microscopy to visualize FOXA1 nuclear organization

      • Live-cell imaging of FOXA1 dynamics

      • In vivo imaging of FOXA1 expression

  • Recombinant antibody fragments with site-specific conjugation:

    • Enhanced properties:

      • Defined antibody-to-dye ratio

      • Reduced batch-to-batch variability

      • Possibility for oriented immobilization

    • Applications:

      • Highly reproducible ChIP-seq experiments

      • Precise protein quantification

      • Multiplexed imaging with minimal cross-reactivity

  • Antibody-oligonucleotide conjugates:

    • Innovative applications:

      • Proximity ligation assays with enhanced specificity

      • DNA-PAINT super-resolution imaging of nuclear FOXA1 organization

      • CITE-seq for combined protein and gene expression profiling

    • Advantages:

      • Single-molecule detection sensitivity

      • Multiplexing capacity through DNA barcoding

      • Integration with spatial transcriptomics

  • Bispecific and multispecific antibodies:

    • Design strategy:

      • Target FOXA1 and interacting partners simultaneously

      • Combine with cell-type specific markers

    • Applications:

      • Detect specific FOXA1 complexes (e.g., FOXA1-AR or FOXA1-ER)

      • Study context-dependent FOXA1 interactions

      • Selective isolation of cell populations with specific FOXA1 interaction states

  • Intrabodies and chromobodies:

    • Innovative approach:

      • Express antibody fragments intracellularly

      • Fuse with fluorescent proteins for live visualization

    • Applications:

      • Track FOXA1 dynamics during development or disease progression

      • Visualize real-time recruitment to chromatin

      • Potentially interfere with specific FOXA1 interactions for functional studies

These advanced antibody technologies will enable unprecedented insights into FOXA1 biology in complex tissue environments, facilitating dynamic, high-resolution studies of this critical transcription factor in development and disease.

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.