HOXB3 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HOXB3 Antibody

HOXB3 (Homeobox B3) antibodies are immunoglobulin-based reagents designed to target the HOXB3 protein, encoded by the HOXB3 gene located on chromosome 17. This protein belongs to the Antennapedia (Antp) homeobox family and regulates anterior-posterior axis development during embryogenesis . In cancer biology, HOXB3 exhibits context-dependent roles, acting as either an oncogene or tumor suppressor depending on tissue type .

Glioblastoma (GBM)

HOXB3 is overexpressed in GBM tissues and cell lines (U87-MG, U251-MG). Knockdown experiments show that HOXB3 silencing:

  • Reduces proliferation by 40–60% in vitro

  • Decreases invasion by downregulating mesenchymal markers (N-cadherin, vimentin) and upregulating E-cadherin

  • Correlates with poor patient survival

Breast Cancer

  • Expression Patterns: HOXB3 is downregulated in hormone receptor-negative subtypes (ER-/PR-), correlating with higher tumor grade and poor prognosis .

  • Mechanisms: Loss of HOXB3 activates pathways linked to immune evasion (e.g., reduced CD8+ T cell infiltration) and cytoskeletal remodeling .

Prostate Cancer

In castration-resistant prostate cancer (CRPC), HOXB3:

  • Drives WNT/β-catenin pathway activation

  • Serves as an independent prognostic marker for PSA progression and mortality

Functional Mechanisms

HOXB3 modulates critical pathways through:

  1. Transcriptional Regulation: Direct binding to Jag1 promoter regions, enhancing Notch signaling in pharyngeal development .

  2. Epithelial-Mesenchymal Transition (EMT): Promotes invasion by suppressing E-cadherin and upregulating vimentin in GBM .

  3. Immune Modulation: Low HOXB3 expression in breast cancer correlates with reduced immune cell infiltration (e.g., B cells, macrophages) .

Research Applications

HOXB3 antibodies enable:

  • Biomarker Discovery: Identifying HOXB3 as a prognostic marker in CRPC and breast cancer .

  • Pathway Analysis: Mapping HOXB3’s role in WNT signaling and matrix adhesion pathways .

  • Therapeutic Targeting: Validating HOXB3 as a potential target in GBM and CRPC .

Future Directions

  1. Clinical Validation: Prospective studies to confirm HOXB3’s utility as a biomarker in CRPC and breast cancer .

  2. Mechanistic Studies: Elucidating HOXB3’s interplay with immune checkpoints (e.g., PD-L1) in tumor microenvironments .

  3. Therapeutic Development: Small-molecule inhibitors targeting HOXB3-WNT axis in metastatic cancers .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timeframes.
Synonyms
Homeo box 2G antibody; Homeo box B3 antibody; Homeobox B3 antibody; Homeobox protein Hox-2.7 antibody; Homeobox protein Hox-2G antibody; Homeobox protein Hox-B3 antibody; Homeobox protein HoxB3 antibody; Hox 2.7 antibody; HOX 2G antibody; HOX2 antibody; Hox2.7 antibody; HOX2G antibody; HOXB3 antibody; HXB3_HUMAN antibody
Target Names
HOXB3
Uniprot No.

Target Background

Function
HOXB3 is a sequence-specific transcription factor that plays a critical role in developmental regulation. It functions as part of a system that assigns specific positional identities to cells along the anterior-posterior axis.
Gene References Into Functions
  1. Research suggests a regulatory circuitry involving miR-375, HOXB3, CDCA3, and DNMT3B that contributes to the development of acute myeloid leukemia (AML). Restoring miR-375 expression may represent a potential therapeutic strategy for AML. PMID: 29439669
  2. HOXB3 is degraded by miR-375 in breast cancer cells. HOXB3 plays a role in tamoxifen resistance. PMID: 28075453
  3. miR-10b may control cell apoptosis, proliferation, migration, and invasion in endometrial cancer by regulating HOXB3 expression. PMID: 27447302
  4. Studies have shown that decreased methylation at HOXB3 and HOXB4 is associated with increased gene expression of both HOXB genes, specifically in mid-risk AML. Conversely, increased DNA methylation at DCC, characteristic of high-risk AML, is linked to increased gene expression. PMID: 25996682
  5. Research has described familial cases of thyroid hormone (TH) deficiency across two generations (proband and his father), alongside two sporadic cases. Polymorphisms in the HOXB3, HOXD3, and PITX2 genes have been identified in these individuals. PMID: 24127533
  6. HoxB3 promotes prostate cancer progression by upregulating CDCA3 expression. PMID: 23219899
  7. HOXb3 expression was found to be weak in patients with inv(7) positive leukemia. PMID: 15674412
  8. RASSF1A silencing strongly correlates with overexpression of HOXB3 and DNMT3B. PMID: 19854132

Show More

Hide All

Database Links

HGNC: 5114

OMIM: 142966

KEGG: hsa:3213

STRING: 9606.ENSP00000308252

UniGene: Hs.654560

Protein Families
Antp homeobox family
Subcellular Location
Nucleus.

Q&A

What is HOXB3 and why is it significant in cancer research?

HOXB3 belongs to the homeobox family of transcription factors that play crucial roles in embryonic development and cellular differentiation. Recent studies have identified HOXB3 as a key player in multiple cancer types, making it an important research target. In castration-resistant prostate cancer (CRPC), HOXB3 protein levels serve as an independent risk factor for PSA progression and patient mortality . Similarly, in glioblastoma (GBM), HOXB3 is highly expressed in cancerous tissues but barely detectable in normal brain tissue, suggesting its potential as a diagnostic or therapeutic target . The significance of HOXB3 lies in its demonstrated abilities to promote tumor cell proliferation, enhance invasiveness, and contribute to treatment resistance, particularly in hormone-refractory cancers. Understanding HOXB3's functions requires reliable antibodies for detection and characterization across experimental systems.

What cellular processes does HOXB3 regulate in cancer progression?

HOXB3 regulates multiple cellular processes that collectively drive cancer progression. In CRPC, HOXB3 functions as a downstream transcription factor in the WNT signaling pathway, where it can transcriptionally regulate multiple WNT pathway genes following nuclear translocation . This activity is particularly significant in the context of APC deficiency, which allows HOXB3 to escape from the destruction complex. In glioblastoma, HOXB3 silencing reduces cell proliferation capacity significantly, with differences becoming apparent by day 3 post-silencing . Furthermore, HOXB3 plays a critical role in tumor cell invasion through modulation of epithelial-mesenchymal transition (EMT). When HOXB3 is silenced in GBM cells, researchers observe decreased expression of mesenchymal markers N-cadherin and vimentin, alongside increased expression of the epithelial marker E-cadherin . This molecular reprogramming corresponds with reduced invasive capacity and morphological changes from elongated, fibroblastic cells to rounded, cobbled cells resembling epithelial morphology.

How do I select the appropriate HOXB3 antibody for my experimental needs?

Selecting the appropriate HOXB3 antibody requires consideration of several critical factors:

  • Epitope targeting: Different antibodies target specific regions of the HOXB3 protein. Available options include antibodies targeting the C-terminus, N-terminus, or middle regions (AA 119-148, 72-121, 108-157, or 315-423) . The choice depends on your experimental question and whether certain domains may be masked in your experimental system.

  • Species reactivity: Verify the antibody's reactivity with your experimental model. Some HOXB3 antibodies show high sequence identity across species (human, bovine: 100%; pig: 93%; mouse, rat: 92%) , but cross-reactivity should be experimentally confirmed.

  • Application compatibility: Ensure the antibody is validated for your specific application:

    • Western blotting

    • Immunohistochemistry (paraffin-embedded or frozen sections)

    • Immunocytochemistry

    • Immunofluorescence

  • Clonality: Polyclonal antibodies (such as those derived from rabbit) offer higher sensitivity through multiple epitope recognition but may have higher batch-to-batch variability compared to monoclonal antibodies.

  • Validation in similar research contexts: Review the literature to identify antibodies successfully employed in experimental systems similar to yours, particularly in the same cancer type or cellular context.

Always perform validation experiments in your specific model system before proceeding with larger studies.

How can I optimize Western blot protocols for HOXB3 detection?

Optimizing Western blot protocols for HOXB3 detection requires attention to several critical parameters:

Sample preparation and protein loading:

  • Extract proteins using RIPA buffer (50 mM Tris-HCl pH 8.0, 1 mM EDTA pH 8.0, 5 mM DTT, 2% SDS) to effectively solubilize nuclear proteins .

  • Determine protein concentration using BCA assay to ensure consistent loading.

  • Load 30 μg of total protein per lane for optimal signal-to-noise ratio when detecting HOXB3 .

  • Include positive control samples with known HOXB3 expression (e.g., U87-MG or U251-MG glioblastoma cells for brain cancer studies) .

Electrophoresis and transfer conditions:

  • Use 10% SDS-PAGE for optimal resolution of HOXB3 (molecular weight ~44 kDa).

  • Transfer to PVDF membranes at controlled voltage to prevent protein loss.

  • Verify transfer efficiency with reversible protein stains before blocking.

Antibody incubation parameters:

  • Block membranes with 5% non-fat dry milk in Tris-buffered saline (pH 7.5) for 1 hour at room temperature.

  • Incubate with HOXB3 primary antibody at recommended dilution (e.g., 1:400 for certain HOXB3 antibodies) overnight at 4°C .

  • Use appropriate HRP-conjugated secondary antibody (typically 1:2,000 dilution) for 1 hour at room temperature .

Signal detection and analysis:

  • Develop using enhanced chemiluminescence (ECL) systems.

  • Normalize HOXB3 signals to appropriate loading controls (β-actin at 1:4,000 dilution has been validated) .

  • Perform densitometry using software like ImageQuant 5.2 for quantitative analysis .

If weak signals persist, optimize by adjusting antibody concentration, extending incubation time, or employing signal amplification systems.

What are the best practices for immunohistochemical detection of HOXB3 in tumor samples?

Successful immunohistochemical detection of HOXB3 in tumor samples requires attention to these methodological details:

Tissue preparation:

  • Fix tissues in formalin and embed in paraffin following standard protocols.

  • Section tissues at 5 μm thickness for optimal antibody penetration and signal resolution .

  • Mount sections on positively charged slides to prevent tissue detachment during antigen retrieval.

Antigen retrieval:

  • Perform heat-induced epitope retrieval by boiling slides in 10 mM sodium citrate buffer (pH 6.0) for 10 minutes .

  • Allow slides to cool gradually in buffer before proceeding to blocking step.

Blocking and antibody incubation:

  • Block non-specific binding with blocking buffer containing 0.2% Triton X-100, 5% normal donkey serum, and 5% non-fat milk in Tris-buffered saline for 1 hour at room temperature .

  • Incubate with primary HOXB3 antibody (dilution 1:200 has been validated) in blocking buffer overnight at 4°C .

  • Wash thoroughly with TBST (Tris-buffered saline containing 0.1% Tween-20).

  • Incubate with alkaline phosphatase-conjugated secondary antibody (1:500) for 1 hour at room temperature .

Signal development and visualization:

  • Develop signal using 3,3-diaminobenzidine tetrahydrochloride (DAB) to produce brown precipitates .

  • Counterstain with hematoxylin (2 minutes at room temperature) to visualize tissue architecture and negatively stained cells .

  • Dehydrate through graded alcohols, clear in xylene, and mount with permanent mounting medium.

Controls and interpretation:

  • Include positive controls (GBM tissue samples show strong HOXB3 staining) and negative controls (normal brain tissue samples show minimal HOXB3 staining) .

  • Assess staining pattern, intensity, and subcellular localization (nuclear vs. cytoplasmic).

  • Quantify expression using appropriate scoring systems based on percentage of positive cells and staining intensity.

How can I validate HOXB3 antibody specificity in my experimental system?

Rigorous validation of HOXB3 antibody specificity is essential for reliable research outcomes. Implement these validation approaches:

Genetic knockdown/knockout controls:

  • Generate stable cell lines with HOXB3 knockdown using lentiviral shRNA technology as demonstrated in U87-MG and U251-MG glioblastoma cells .

  • Compare antibody reactivity between parental cells and knockdown cells via Western blot and immunocytochemistry.

  • A significant reduction in signal intensity in knockdown cells confirms antibody specificity for HOXB3.

Peptide competition assay:

  • Pre-incubate the HOXB3 antibody with excess immunizing peptide (the synthetic peptide used to generate the antibody) .

  • Perform parallel experiments with blocked and unblocked antibody.

  • Disappearance of signal in the peptide-blocked condition confirms specific binding to the target epitope.

Multi-antibody approach:

  • Test multiple antibodies targeting different epitopes of HOXB3 (N-terminal, C-terminal, and middle regions are available) .

  • Concordant results from antibodies recognizing different epitopes strengthen confidence in specificity.

Cross-species reactivity assessment:

  • If your antibody claims cross-reactivity (e.g., human/mouse/rat), validate this experimentally in each species of interest.

  • While sequence identity predictions suggest reactivity (92-100% identity across mammals), functional validation is necessary .

Mass spectrometry validation:

  • For definitive validation, immunoprecipitate HOXB3 using your antibody and confirm protein identity via mass spectrometry.

  • This approach identifies both the target protein and potential cross-reactive proteins.

How should I design experiments to investigate HOXB3's role in cancer cell invasion?

Investigating HOXB3's role in cancer cell invasion requires a systematic experimental approach:

Baseline expression analysis:

  • Quantify HOXB3 expression across multiple cancer cell lines and normal tissue controls using both qRT-PCR and Western blotting.

  • Select high-expressing cell lines (e.g., U87-MG and U251-MG for glioblastoma studies) for manipulation experiments .

Genetic manipulation strategies:

  • Establish stable HOXB3 knockdown cell lines using lentiviral shRNA delivery systems with appropriate puromycin selection .

  • Generate HOXB3 overexpression models in low-expressing cell lines.

  • Create inducible expression systems to study temporal effects of HOXB3 modulation.

Invasion assay design:

  • Perform Matrigel invasion assays using identical cell numbers from parental and HOXB3-manipulated cells.

  • Incubate cells for 36 hours to allow sufficient time for invasion to occur .

  • Quantify invaded cells through counting of multiple representative fields.

  • Include technical and biological replicates for statistical validation.

Molecular mechanism investigation:

  • Assess EMT marker expression (E-cadherin, N-cadherin, vimentin) via Western blotting in response to HOXB3 modulation .

  • Evaluate morphological changes using phase-contrast microscopy (transition from elongated fibroblastic to rounded epithelial morphology) .

  • Investigate cellular adhesion properties through adhesion assays to various extracellular matrix components.

Advanced mechanistic studies:

  • Perform RNA-sequencing in HOXB3-negative versus HOXB3-high tumors to identify regulated pathways, as done in CRPC studies .

  • Assess WNT pathway activation through TOPFlash reporter assays and β-catenin localization studies .

  • Investigate transcriptional targets of HOXB3 using ChIP-seq approaches.

This comprehensive experimental design allows for both phenotypic characterization and molecular mechanism elucidation of HOXB3's role in cancer cell invasion.

What controls are essential when studying HOXB3's relationship with the WNT signaling pathway?

Investigating HOXB3's interaction with the WNT signaling pathway requires rigorous controls to establish causality and specificity:

Essential controls for WNT-HOXB3 interaction studies:

Control TypeImplementationRationale
Genetic controlsHOXB3 knockdown/knockout linesDemonstrate HOXB3 requirement
APC-deficient cell modelsTest HOXB3 dependency on APC status
WNT3A knockdown/knockout linesValidate upstream regulation of HOXB3
Pharmacological controlsWNT pathway inhibitors (e.g., tankyrase inhibitors)Confirm pathway specificity
Dose-response experimentsEstablish concentration-dependent effects
Time-course experimentsDetermine temporal dynamics
Cellular localization controlsCytoplasmic/nuclear fractionationTrack HOXB3 translocation
Immunofluorescence with compartment markersVisualize subcellular distribution
Functional readoutsWNT reporter assays (TOPFlash)Measure canonical WNT activity
β-catenin stabilization assaysConfirm pathway activation
Target gene expression (AXIN2, LGR5, etc.)Validate downstream effects

Mechanistic controls for destruction complex studies:

  • Compare HOXB3 behavior in APC-proficient versus APC-deficient cells to evaluate destruction complex involvement .

  • Assess HOXB3-APC interaction through co-immunoprecipitation experiments with appropriate antibody controls.

  • Evaluate HOXB3 phosphorylation status in response to GSK3β inhibition/activation.

Treatment response controls:

  • Compare abiraterone responsiveness in APC-deficient CRPC xenografts with normal versus suppressed HOXB3 expression .

  • Include time-matched untreated controls to account for changes in tumor biology over time.

  • Monitor both short-term (biochemical) and long-term (phenotypic) responses to establish causality.

Implementing these controls will establish whether HOXB3 functions as a downstream effector of WNT signaling, a parallel pathway component, or an upstream regulator in your specific cancer model.

How can I reconcile conflicting data regarding HOXB3 function in different cancer types?

Reconciling conflicting data on HOXB3 function across cancer types requires systematic analysis and experimental validation:

Context-dependent functions of HOXB3:
Research has revealed that HOXB3 can exhibit seemingly contradictory functions in different cancers. For example, in glioblastoma, HOXB3 silencing inhibits cell invasion , whereas in pancreatic cancer, HOXB3 silencing has been reported to reduce migration . In colorectal cancer, HOXB3 appears to regulate proliferation without affecting invasion . These discrepancies may reflect genuine biological differences rather than experimental artifacts.

Methodological approaches to resolve contradictions:

  • Perform parallel experiments across multiple cancer models:

    • Implement identical HOXB3 manipulation protocols in different cancer cell lines simultaneously.

    • Use consistent assay conditions, reagents, and quantification methods.

    • This approach controls for technical variables that might contribute to apparent contradictions.

  • Investigate cellular context determinants:

    • Characterize baseline expression of HOXB3 cofactors and interacting proteins across models.

    • Assess expression of competing or redundant HOX family members.

    • Evaluate the activation status of signaling pathways that might interact with HOXB3 (e.g., WNT, AR).

  • Examine cancer-specific protein interactions:

    • Perform immunoprecipitation followed by mass spectrometry to identify HOXB3 binding partners in each cancer type.

    • Compare these interactomes to identify cancer-specific cofactors that might redirect HOXB3 function.

  • Assess post-translational modifications:

    • Investigate cancer-specific phosphorylation, acetylation, or other modifications of HOXB3.

    • These modifications could alter protein stability, localization, or transcriptional activity.

  • Evaluate genomic binding profiles:

    • Perform ChIP-seq in different cancer types to determine if HOXB3 binds distinct genomic loci.

    • Integrate with RNA-seq data to connect binding patterns with differential gene regulation.

  • Create domain-specific mutants:

    • Generate HOXB3 constructs with mutations in specific functional domains.

    • Test these in different cancer models to identify domain requirements for context-specific functions.

By implementing these approaches, researchers can determine whether HOXB3's apparently contradictory functions reflect true biological differences in cancer-specific molecular contexts or arise from methodological variations across studies.

How can HOXB3 antibodies be used to stratify patients for targeted therapies?

HOXB3 expression may serve as a biomarker for patient stratification, particularly in treatment response prediction:

Patient stratification methodology using HOXB3 antibodies:

The development of companion diagnostic tests using HOXB3 antibodies could ultimately guide treatment selection, particularly for patients with advanced cancers where resistance mechanisms are critical determinants of outcome.

What methodological approaches can identify transcriptional targets of HOXB3 in cancer cells?

Identifying the transcriptional targets of HOXB3 requires integrated genomic approaches:

Chromatin immunoprecipitation sequencing (ChIP-seq):

  • Utilize validated HOXB3 antibodies to immunoprecipitate HOXB3-bound chromatin.

  • Include appropriate controls: input chromatin, IgG control, and HOXB3-knockdown cells.

  • Perform in multiple cell lines to identify context-dependent and core binding sites.

  • Analyze binding motifs to determine direct HOXB3 binding sequences versus co-factor mediated binding.

RNA sequencing after HOXB3 modulation:

  • Compare transcriptomes between HOXB3-high and HOXB3-negative tumors, as demonstrated in CRPC studies .

  • Perform time-course RNA-seq after inducible HOXB3 activation to distinguish primary from secondary effects.

  • Apply pathway enrichment analysis to identify coordinated gene programs (e.g., WNT pathway genes) .

Integration of ChIP-seq with RNA-seq:

  • Correlate HOXB3 binding sites with differentially expressed genes.

  • Focus on genes with both HOXB3 binding and expression changes for validation studies.

  • Utilize computational approaches to reconstruct transcriptional networks.

Functional validation of direct targets:

  • Design reporter constructs containing putative HOXB3-binding promoter regions.

  • Perform site-directed mutagenesis of binding motifs to confirm direct regulation.

  • Validate individual targets through qRT-PCR, Western blotting, and functional assays.

This integrated approach has successfully identified WNT pathway genes as HOXB3 targets in CRPC and could be applied to identify context-specific targets in other cancer types.

How can I troubleshoot non-specific binding of HOXB3 antibodies in immunohistochemistry?

Non-specific binding in HOXB3 immunohistochemistry can be addressed through systematic optimization:

Common causes and solutions for non-specific binding:

ProblemPotential CausesSolutions
High background stainingInsufficient blockingExtend blocking time to 2 hours; try alternative blocking agents (BSA, serum)
Excessive antibody concentrationPerform antibody titration series (1:100 to 1:1000)
Secondary antibody cross-reactivityUse secondary antibodies pre-adsorbed against host species proteins
Non-nuclear stainingEpitope maskingTest alternative antigen retrieval methods (citrate vs. EDTA buffers)
Antibody specificity issuesCompare staining patterns with antibodies targeting different HOXB3 epitopes
Fixation artifactsStandardize fixation protocols (time, temperature, buffer composition)
Inconsistent stainingTissue heterogeneityIncrease sampling (multiple cores/sections per specimen)
Batch effectsProcess all comparative samples in the same experimental run
Antibody lot variationRecord lot numbers and test new lots against reference samples

Validation controls for troubleshooting:

  • Process HOXB3-positive (GBM tissue) and HOXB3-negative (normal brain) controls alongside test samples .

  • Include peptide competition controls to identify non-specific binding.

  • Perform parallel staining with multiple antibodies targeting different HOXB3 epitopes .

Optimization of detection systems:

  • Compare chromogenic (DAB) versus fluorescent detection methods.

  • Consider tyramide signal amplification for low-abundance targets.

  • Optimize counterstaining intensity to maintain visibility of specific signals.

Systematic evaluation of these variables will help establish reliable HOXB3 immunohistochemistry protocols with minimal non-specific binding.

What are the most common pitfalls in quantifying HOXB3 expression changes?

Accurate quantification of HOXB3 expression changes requires awareness of several methodological pitfalls:

Western blot quantification challenges:

  • Saturation of signal: Ensure exposure times capture signals within the linear dynamic range.

  • Incomplete transfer of proteins: Verify transfer efficiency with reversible total protein stains.

  • Inappropriate normalization: Select loading controls not affected by experimental conditions.

  • Batch effects: Include common reference samples across multiple blots for inter-blot normalization.

qRT-PCR quantification issues:

  • Primer specificity: Design primers spanning exon-exon junctions to prevent genomic DNA amplification.

  • Reference gene selection: Validate stability of reference genes across experimental conditions using algorithms like geNorm.

  • PCR inhibition: Include internal amplification controls to detect inhibitory contaminants.

  • Threshold setting: Use consistent threshold determination methods across experiments.

Immunohistochemistry quantification challenges:

  • Subjective scoring: Implement digital image analysis for objective quantification.

  • Heterogeneous expression: Assess multiple fields/regions to capture spatial heterogeneity.

  • Threshold determination: Use positive and negative controls to establish scoring thresholds.

  • Inter-observer variability: Have multiple trained observers score samples independently.

Single-cell analysis considerations:

  • Population averaging: Consider single-cell approaches (flow cytometry, single-cell RNA-seq) to detect subpopulations.

  • Rare cell populations: Employ enrichment strategies for cells of interest before analysis.

  • Technical dropouts: Include spike-in controls to assess technical variation in single-cell approaches.

Awareness of these pitfalls and implementation of appropriate controls will enhance the reliability of HOXB3 expression quantification across experimental platforms.

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.