HOXB13 Human

Homeobox B13 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Functional Domains

HOXB13 is a 284-amino acid protein with three critical domains:

  • Homeodomain (216–275 AA): DNA-binding region conserved across species

  • MEIS Interaction Domains: Regulate protein-DNA interactions

  • HoxA13 N-terminal Domain: Involved in transcriptional regulation

Key Pathogenic Mutations

MutationPopulation FrequencyCancer Risk (OR)Functional Impact
G84E0.1% (European)10–20× Disrupts MEIS interactions
R217CRareNot significantUnclear pathogenicity
G135E1.2% (Chinese)3.5× Increased protein stability

Mechanisms of Oncogenesis

  • Loss of Tumor Suppression: Mutations impair HOXB13's ability to regulate cell proliferation .

  • Androgen Receptor (AR) Interaction: HOXB13 modulates AR activity, influencing prostate cancer progression .

  • Epigenetic Dysregulation: Altered HOXB13 expression correlates with DNA hypermethylation in renal cell carcinoma .

Role in Prostate Cancer

HOXB13 is a key player in prostate carcinogenesis:

  • Familial Risk: G84E carriers have 60–70% lifetime prostate cancer risk vs. 12% in non-carriers .

  • Therapeutic Resistance: HOXB13-AR complexes promote castration-resistant prostate cancer by activating oncogenes like RFX6 .

  • Biomarker Potential: HOXB13 expression ratios predict recurrence in ER+ breast cancer .

Clinical Implications Table

ApplicationMechanismStudy Outcome
Risk StratificationGermline mutation screeningEarly detection in high-risk men
Therapeutic TargetHOXB13-AR-V7 complex inhibitionReduced tumor growth in vitro
Prognostic MarkerHOXB13/IL17BR expression ratioPredicts tamoxifen resistance

Computational and Functional Insights

Recent studies employ advanced modeling to characterize HOXB13:

  • Structural Modeling: Full-length HOXB13 simulations reveal mutation-induced stability changes (e.g., G84E increases stability by 1.2 kcal/mol) .

  • SNP Analysis: 21/95 missense SNPs in non-homeobox regions are deleterious, affecting DNA binding .

  • 3′UTR Variants: rs563065128 disrupts stem cell renewal by altering Mushashi binding elements .

Controversies and Emerging Roles

While HOXB13 is firmly linked to prostate cancer, its role in other cancers remains debated:

  • Breast Cancer: No significant association with G84E (OR = 1.1, p = 0.79) .

  • Renal Cell Carcinoma: HOXB13 acts as a tumor suppressor via DNMT3B-mediated methylation .

  • Dual Functions: HOXB13 exhibits both oncogenic and tumor-suppressive effects depending on cellular context .

Research Gaps and Future Directions

  1. Mechanistic Studies: How do HOXB13-MEIS interactions precisely modulate transcription?

  2. Therapeutic Development: Can small molecules disrupt HOXB13-AR complexes in castration-resistant cancer?

  3. Population-Specific Variants: Why do G135E mutations predominantly affect Chinese populations?

Product Specs

Introduction
HOXB13, a transcription factor belonging to the highly conserved homeobox gene family, plays a crucial role in vertebrate embryonic development. This gene is involved in fetal skin development and cutaneous regeneration. Studies in mice have shown that a similar gene exhibits temporal and spatial colinearity along the embryo's main body axis but not in secondary axes, suggesting its involvement in body patterning along the primary axis.
Description
Recombinant human HOXB13, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 307 amino acids (residues 1-284) with a molecular weight of 33.1 kDa. This protein is expressed with a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear solution, sterilized by filtration.
Formulation
HOXB13 protein is supplied in a solution at a concentration of 0.5 mg/ml. The solution contains 20 mM Tris-HCl buffer (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep the vial refrigerated at 4°C. For extended storage, freeze the solution at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 85.0% as determined by SDS-PAGE analysis.
Synonyms
Homeobox B13, Homeobox Protein Hox-B13, PSGD.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMEPGNYA TLDGAKDIEG LLGAGGGRNL VAHSPLTSHP AAPTLMPAVN YAPLDLPGSA EPPKQCHPCP GVPQGTSPAP VPYGYFGGGY YSCRVSRSSL KPCAQAATLA AYPAETPTAG EEYPSRPTEF AFYPGYPGTY QPMASYLDVS VVQTLGAPGE PRHDSLLPVD SYQSWALAGG WNSQMCCQGE QNPPGPFWKA AFADSSGQHP PDACAFRRGR KKRIPYSKGQ LRELEREYAA NKFITKDKRR KISAATSLSE RQITIWFQNR RVKEKKVLAK VKNSATP

Q&A

What is HOXB13 and what role does it play in normal prostate development?

HOXB13 is a homeobox transcription factor that serves as a key lineage factor critical for prostate gland differentiation. In murine models, HOXB13 expression increases during later stages of prostate development, suggesting its importance in terminal differentiation processes . During embryonic development, HOX genes including HOXB13 show specific spatiotemporal expression patterns that regulate proper organ formation. In human prostatic development, HOXB13 works in concert with other HOX proteins, particularly those from the HOXA and HOXD clusters, to ensure proper prostatic epithelial differentiation . The protein functions primarily in the nucleus, as confirmed by immunohistochemical staining showing nuclear localization in both murine and human prostatic epithelial cells .

How does HOXB13 expression differ across species in prostate development?

There are notable species-specific differences in HOX gene expression patterns during prostate development. In mice, Hoxa10 expression peaks at embryonic day 19 (E19) and decreases rapidly after birth, while Hoxa13 and Hoxd13 peak around E15 and gradually diminish into adulthood . In contrast, rat prostates show a postnatal increase in Hoxa13, Hoxd13, and Hoxb13 expression that persists into adulthood . Spatial expression also differs: in mice, Hoxa13 and Hoxd13 expression peaks in the seminal vesicle, while in rats, these genes show highest expression in the dorsal prostate with clear anterior boundaries at the epididymis . In adult human tissues, HOXA9, HOXA11, HOXA13, HOXB13, and HOXD9 are identified as the highest expressing HOX genes in the prostate, demonstrating that while similar HOX genes are expressed across species, the timing, location, and expression levels vary significantly .

What are the validated methods for detecting HOXB13 protein expression in tissues?

Multiple validated techniques exist for detecting HOXB13 protein expression in various sample types. For western blot analysis, sheep anti-human HOXB13 antigen affinity-purified polyclonal antibody has been validated using genetic controls and successfully detects HOXB13 at approximately 34 kDa in prostate cancer cell lines like LNCaP . For cellular localization studies, immunofluorescence using antibodies such as the NorthernLights™ 557-conjugated anti-sheep IgG secondary antibody following primary HOXB13 antibody application has demonstrated specific nuclear staining . For tissue sections, formalin-fixed paraffin-embedded samples can be analyzed using immunohistochemistry with HRP-DAB detection systems and hematoxylin counterstaining, which effectively visualizes HOXB13 in epithelial cell nuclei . Proper validation using genetic controls is essential to ensure specificity, as demonstrated in recent studies that have rigorously tested HOXB13 antibodies using appropriate positive and negative controls .

What is the G84E mutation and how significantly does it increase prostate cancer risk?

The G84E mutation (c.251G>A) in the HOXB13 gene represents a glycine to glutamic acid substitution at position 84. This germline mutation significantly increases prostate cancer risk, with relative risk estimates ranging from 3.3 to 20.1-fold across different population studies . In the initial discovery study by Ewing et al., the mutation conferred an odds ratio of 20.1 (95% CI: 3.5-803.3) for prostate cancer risk . Subsequent replication studies found consistent but somewhat lower risk estimates: Stott-Miller et al. reported a 3.3-fold relative risk (95% CI: 1.2-9.0), while Karlsson et al. found a 3.4-fold increased risk (95% CI: 2.2-5.4) in a large Swedish cohort . The risk is particularly elevated in men with family history of prostate cancer, with some family-based studies showing odds ratios as high as 68.1 . Additionally, the G84E variant appears to be associated with earlier age of diagnosis, with one study showing a carrier frequency of 10.3% among cases diagnosed between ages 35-55 years (OR=8.6, 95% CI: 5.1-14.0) .

What ethnic variations exist in HOXB13 mutations associated with prostate cancer?

Significant ethnic variations exist in HOXB13 mutations associated with prostate cancer risk, with different populations harboring distinct variants. The G84E mutation has been primarily identified in populations of European/Caucasian descent and is rare or absent in other ethnicities . For instance, in Chinese populations, researchers identified a novel G135E mutation associated with increased prostate cancer risk, while the G84E mutation was not detected . Similarly, in Portuguese men, the A128D and F240L mutations were identified as risk variants . In African or African-American populations, rare variants such as R229G and G216C have been reported, though their clinical significance requires further investigation . The Chen et al. study explicitly noted that the G84E allele was detected only in Caucasians within their multi-ethnic cohort . These findings highlight the importance of conducting genetic studies across diverse populations, as different ethnic groups may harbor distinct mutations in the same gene that confer similar cancer risks.

How does HOXB13 expression change across the spectrum of prostate cancer progression?

HOXB13 expression demonstrates dynamic changes across the spectrum of prostate cancer progression. In localized prostate cancer, all tumors show detectable HOXB13 protein expression, though interestingly, lower HOXB13 expression levels are observed in higher-grade tumors . Despite this observation, studies have not found significant associations between HOXB13 expression levels and disease recurrence or disease-specific survival in localized disease . As prostate cancer progresses to metastatic castration-resistant disease, HOXB13 expression is retained in the majority of tumors, making it a useful diagnostic marker even in advanced disease stages . In tumors showing evidence of lineage plasticity, lower HOXB13 protein and mRNA levels are observed, but remarkably, 84% of androgen receptor-negative castration-resistant prostate cancers and neuroendocrine prostate cancers still retain detectable levels of HOXB13 . This retention of HOXB13 expression even in advanced and dedifferentiated tumors suggests a fundamental role for this transcription factor in maintaining prostatic lineage identity even under selective pressures that alter other lineage markers.

What is the relationship between HOXB13 expression and androgen receptor signaling in prostate cancer?

HOXB13 and androgen receptor (AR) signaling demonstrate a complex interrelationship in prostate cancer. While both are critical lineage-specific factors in prostatic epithelium, their expression patterns can diverge during disease progression. Notably, even in androgen receptor-negative castration-resistant prostate cancers, 84% retain detectable levels of HOXB13, indicating that HOXB13 expression can be maintained through AR-independent mechanisms . This finding is particularly significant for tracking tumor lineage in the setting of treatment resistance, where AR signaling may be lost or altered. The persistence of HOXB13 expression when AR expression is lost suggests that HOXB13 may represent a more fundamental and stable marker of prostatic origin. Research indicates that HOXB13 can function as both a coactivator and corepressor of AR in a context-dependent manner, influencing the transcriptional programs that drive prostate cancer progression . This complex relationship may contribute to the diverse phenotypes observed in advanced prostate cancers and suggests that targeting HOXB13 might provide therapeutic opportunities even in AR-independent disease contexts.

What epigenetic mechanisms regulate HOXB13 expression in prostate cancer?

Epigenetic mechanisms play a significant role in regulating HOXB13 expression in prostate cancer, particularly in advanced disease states. In neuroendocrine prostate cancers (NEPCs), the reduced expression of HOXB13 has been specifically associated with a gain of CpG methylation in the HOXB13 gene body . This epigenetic modification can lead to transcriptional repression, explaining the decreased expression observed in some dedifferentiated tumors. DNA methylation represents just one aspect of the epigenetic regulation of HOXB13; histone modifications likely also contribute to its expression control, though these mechanisms are less well-characterized in the current literature. Unlike genetic mutations that permanently alter gene function, epigenetic changes are potentially reversible, suggesting possibilities for therapeutic interventions that might restore normal HOXB13 expression patterns. The identification of specific epigenetic mechanisms regulating HOXB13 provides insight into how prostatic lineage identity can be maintained or lost during cancer progression and may help explain the heterogeneity observed in advanced prostate cancers.

What are the validated antibodies and protocols for HOXB13 immunohistochemistry in research?

Several validated antibodies and protocols have been established for HOXB13 immunohistochemistry (IHC) with proven reliability in research settings. The sheep anti-human HOXB13 antigen affinity-purified polyclonal antibody (e.g., R&D Systems catalog #AF8156) has been rigorously validated using genetic controls and is effective for IHC applications at concentrations of 1-10 μg/mL . For optimal results in formalin-fixed paraffin-embedded (FFPE) tissue sections, incubation with primary antibody overnight at 4°C followed by detection using an HRP-DAB system (such as Anti-Sheep HRP-DAB Cell & Tissue Staining Kit) and hematoxylin counterstaining yields specific nuclear staining in prostatic epithelial cells . Critical validation steps include the use of appropriate positive controls (e.g., normal prostate tissue) and negative controls (tissues known to lack HOXB13 expression or genetically engineered HOXB13-knockout samples) . For fluorescent IHC, protocols using NorthernLights™ 557-conjugated secondary antibodies with DAPI counterstaining effectively demonstrate nuclear localization . Researchers should be aware that antigen retrieval methods, antibody concentrations, and incubation conditions may require optimization for specific tissue types and fixation methods.

How can researchers effectively detect and verify novel HOXB13 mutations in diverse populations?

Effective detection and verification of novel HOXB13 mutations in diverse populations requires a multi-faceted approach combining various genomic and confirmatory techniques. Initial screening can be performed using targeted next-generation sequencing of the HOXB13 gene, focusing on the coding regions and splice junctions where most pathogenic mutations occur . This should be followed by Sanger sequencing for verification of potential variants. Population-specific approaches are critical—researchers should design studies with adequate representation of diverse ethnic groups, as demonstrated by studies that identified the G135E mutation in Chinese populations and A128D/F240L mutations in Portuguese cohorts . For novel variants, segregation analysis in families can provide evidence of pathogenicity, as was initially done for the G84E mutation . Functional studies using site-directed mutagenesis to introduce identified mutations into HOXB13 expression constructs, followed by transcriptional reporter assays, protein-protein interaction studies, and subcellular localization experiments can help characterize the biological impact of novel variants . Case-control studies with appropriate ethnic matching are essential to establish population-specific risk estimates for newly identified variants. Researchers should also consider whole-genome or whole-exome sequencing approaches to capture the full spectrum of variation in and around the HOXB13 gene.

What experimental models are available for studying HOXB13 function in prostate development and cancer?

A diverse array of experimental models is available for investigating HOXB13 function in prostate development and cancer. Cell line models include established prostate cancer lines such as LNCaP, which express detectable levels of HOXB13 and can be used for in vitro studies of transcriptional regulation, protein interactions, and cellular phenotypes following HOXB13 manipulation . For genetic manipulation, CRISPR/Cas9 gene editing can generate HOXB13 knockout or knockin models with specific mutations of interest. Animal models include transgenic mice with prostate-specific expression of wild-type or mutant HOXB13, as well as knockout models to study developmental consequences of HOXB13 loss . Xenograft models using patient-derived tissues can maintain the complex cellular architecture of prostate tumors while allowing experimental manipulation of HOXB13 expression or function. Organoid cultures derived from normal prostate or prostate cancer tissues provide three-dimensional systems that better recapitulate tissue architecture while remaining amenable to genetic manipulation. For developmental studies, embryonic stem cell differentiation protocols can model the role of HOXB13 during prostate specification and maturation. Each model system offers distinct advantages for addressing specific research questions, from molecular mechanisms to tissue-level phenotypes.

How does HOXB13 compare to other prostate lineage markers for diagnostic applications?

HOXB13 demonstrates superior performance compared to other prostate lineage markers for diagnostic applications, particularly in advanced disease states. In a comprehensive comparative study, HOXB13 showed greater sensitivity than the commonly used prostate lineage marker NKX3.1 in detecting advanced metastatic prostate cancers . When evaluated in a large cohort of 837 patients (383 with prostatic tumors and 454 with non-prostatic tumors), HOXB13 immunohistochemistry demonstrated remarkably high sensitivity (97%) and specificity (99%) for prostatic origin . This exceptional diagnostic performance is particularly valuable in the context of metastatic disease, where determining the tissue of origin is critical for treatment decisions. Unlike prostate-specific antigen (PSA) and other androgen-regulated markers that may be lost in treatment-resistant disease, HOXB13 expression persists in the majority of cases, including 84% of androgen receptor-negative tumors . This stability makes HOXB13 particularly valuable for tracking prostatic lineage throughout disease progression and treatment. Additionally, the nuclear localization of HOXB13 provides a clear and easily interpretable staining pattern compared to some cytoplasmic or membranous markers.

What is the mechanistic role of HOXB13 mutations in prostate carcinogenesis?

The mechanistic role of HOXB13 mutations in prostate carcinogenesis remains incompletely understood, though several pathways have been implicated. The G84E mutation occurs in a highly conserved region of the protein that is involved in protein-protein interactions, suggesting it may alter HOXB13's ability to interact with transcriptional cofactors or DNA . As a transcription factor, mutant HOXB13 likely causes dysregulation of target genes involved in cellular proliferation, differentiation, and hormone signaling. The relationship between HOXB13 and androgen receptor (AR) signaling is particularly relevant, as HOXB13 can modulate AR-dependent transcription and may contribute to altered hormone responses in prostate tissue . Different mutations (G84E, G135E, A128D, etc.) occur in distinct protein domains and may have varying functional consequences, which could explain population-specific effects and varying penetrance . While HOXB13 mutations clearly increase prostate cancer risk, they may function as "genetic modifiers" rather than classical tumor suppressors or oncogenes, potentially explaining why the mutations are associated with increased risk but are not universally present in all tumors from carrier families . Further mechanistic studies using isogenic cell lines, patient-derived models, and transgenic animals are needed to fully elucidate how HOXB13 mutations promote carcinogenesis.

What therapeutic implications arise from understanding HOXB13 biology in prostate cancer?

Understanding HOXB13 biology in prostate cancer opens several promising therapeutic avenues. As a lineage-specific transcription factor that persists even in treatment-resistant disease, HOXB13 represents a potential therapeutic target that could be effective regardless of androgen receptor status . Transcription factors have traditionally been considered "undruggable," but advances in targeted protein degradation (e.g., PROTACs, molecular glues) and allosteric modulators may overcome this limitation. The high specificity of HOXB13 for prostatic tissue (99% specificity) suggests that targeting it could achieve selective effects on prostate cancer with minimal impact on other tissues . For carriers of HOXB13 mutations, enhanced surveillance protocols could be developed for early detection, similar to approaches used for other hereditary cancer syndromes. Understanding the downstream pathways regulated by HOXB13 may identify more easily targetable vulnerabilities in HOXB13-driven cancers. The epigenetic regulation of HOXB13, particularly the observation of gene body methylation in neuroendocrine prostate cancers, suggests that epigenetic therapies might modulate HOXB13 expression and potentially restore sensitivity to conventional treatments . Additionally, the persistence of HOXB13 expression in metastatic disease makes it an attractive target for antibody-drug conjugates or radioimmunoconjugates that could deliver cytotoxic payloads specifically to prostate cancer cells regardless of their location or differentiation state.

What is the performance of HOXB13 as a tissue-of-origin marker for metastatic tumors?

HOXB13 demonstrates excellent performance as a tissue-of-origin marker for metastatic tumors of prostatic origin. In a comprehensive evaluation of 837 patients (383 with prostatic tumors and 454 with non-prostatic tumors), HOXB13 immunohistochemistry achieved a remarkable 97% sensitivity and 99% specificity for prostatic origin . This exceptional performance makes HOXB13 one of the most accurate tissue-specific markers available for diagnostic pathology. Compared to traditional prostate markers, HOXB13 shows greater sensitivity in detecting advanced metastatic prostate cancers than the commonly used lineage marker NKX3.1 . A particularly valuable characteristic of HOXB13 as a diagnostic marker is its persistence in treatment-resistant disease—84% of androgen receptor-negative castration-resistant prostate cancers and neuroendocrine prostate cancers retain detectable levels of HOXB13 . This persistence allows for accurate identification of prostatic origin even in highly dedifferentiated tumors that have lost expression of androgen-regulated markers like PSA. The nuclear localization of HOXB13 provides a clear and easily interpretable staining pattern for pathologists. These characteristics collectively position HOXB13 as a superior tissue-of-origin marker for metastatic tumors, particularly in challenging cases where treatment resistance or dedifferentiation has altered the expression of other prostatic markers.

Product Science Overview

Structure and Function

HOXB13 is characterized by the presence of a homeobox, a specific DNA sequence that encodes a protein domain known as the homeodomain. The homeodomain typically consists of 60 amino acids and is responsible for binding to DNA and regulating gene expression. HOXB13 is involved in the development of various tissues, including the skin and prostate .

Role in Development

HOXB13 has been implicated in fetal skin development and cutaneous regeneration. In mice, a similar gene has shown temporal and spatial colinearity in the main body axis of the embryo, suggesting its role in body patterning along the axis . However, it is not expressed in the secondary axes, indicating specific functions in primary body axis development.

Recombinant HOXB13

Recombinant HOXB13 is a denatured protein with an N-terminal His-tag, corresponding to the amino acids 1-284 of human HOXB13. It is expressed in E. coli and is used for research purposes. The recombinant protein is typically used in studies to understand the function and regulation of HOXB13 in various biological processes .

Applications in Research

The recombinant HOXB13 protein is valuable in research focused on developmental biology, gene regulation, and disease mechanisms. It is particularly useful in studying the molecular pathways involved in skin development and regeneration, as well as its role in prostate cancer. Researchers use this protein to investigate how HOXB13 interacts with other proteins and DNA to regulate gene expression.

Storage and Handling

Recombinant HOXB13 is stored at 4°C for short-term use and at -20°C for long-term storage. It is important to avoid freeze-thaw cycles to maintain the protein’s stability and functionality. The protein is typically supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4M urea .

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 2024 Thebiotek. All Rights Reserved.