Role in Meiotic Recombination: HUS1B participates in alternative 9-1-1 complexes (e.g., RAD1-RAD9B-HUS1B) essential for homolog synapsis, double-strand break (DSB) repair, and meiotic silencing in germ cells .
Checkpoint Activation: These complexes facilitate ATR kinase activation, ensuring proper chromosome segregation .
Therapeutic Target Potential: While HUS1B itself is not directly linked to cancer, its homolog HUS1 has been implicated as an oncogene in urothelial carcinoma (UC), influencing cisplatin resistance .
Complex Dynamics: HUS1B-containing complexes operate independently of canonical 9-1-1 components (RAD9, HUS1), enabling RAD1 to localize to unsynapsed chromatin during meiosis .
Cisplatin Resistance: In UC, HUS1 knockdown sensitizes cisplatin-sensitive cells but fails to reverse resistance in already-resistant cells, suggesting context-dependent roles .
| Component | Canonical Complex Role | Alternative Complex Role |
|---|---|---|
| HUS1 | DSB repair, germ cell development | N/A |
| HUS1B | N/A | Meiotic ATR activation, homolog synapsis |
| RAD1 | Checkpoint signaling | Binds HUS1B/RAD9B in meiosis |
HUS1B is most closely related to HUS1, a component of a cell cycle checkpoint protein complex involved in cell cycle arrest in response to DNA damage. While sharing structural similarities, HUS1B exhibits distinct functional characteristics. Unlike HUS1, HUS1B can interact with the checkpoint protein RAD1 but not with RAD9. Notably, overexpression of HUS1B has been shown to induce cell death, suggesting a related but functionally distinct role compared to HUS1 .
HUS1B has the following molecular identifiers:
UniProt ID: Q8NHY5 (Human)
Entrez Gene ID: 135458 (Human), 691382 (Rat)
Protein aliases include: Checkpoint protein HUS1B, hHUS1B, HUS1 checkpoint homolog b, MGC126746, MGC126748, RP11-532F6.1
HUS1B antibodies are primarily utilized in Western Blot (WB) and ELISA applications in research settings . When comparing to the related HUS1 antibodies, which have broader application potential including Immunohistochemistry (IHC) and Immunofluorescence (IF), researchers should note that HUS1B antibodies currently have more limited validated applications . This difference in application range reflects both the distinct properties of these proteins and the current state of antibody development for HUS1B.
The evidenced applications for HUS1B antibodies include:
| Application | Validation Status |
|---|---|
| Western Blot (WB) | Validated |
| ELISA | Validated |
| Species Reactivity | Human, Rat |
HUS1B expression shows a distinctive pattern across tissues and cell types, though less extensively characterized than HUS1. According to the Pharos database, HUS1B has a knowledge value of 0.56 (on a 0-1 scale) regarding cell type or tissue expression patterns . This indicates moderate but not comprehensive knowledge of its expression profile.
Key knowledge metrics for HUS1B include:
| Knowledge Category | Value (0-1 scale) |
|---|---|
| Tissue sample | 0.81 |
| Cell type or tissue | 0.56 |
| Cellular component | 0.53 |
| Biological process | 0.51 |
| Cell line | 0.50 |
When designing experiments, researchers should consult resources such as The Human Protein Atlas to identify appropriate positive control cell lines that express HUS1B . This step is crucial for establishing experimental validity before proceeding with studies on your cell line of interest.
When designing a flow cytometry experiment to detect HUS1B, researchers must first determine the subcellular localization of HUS1B to select appropriate cell preparation methods. As HUS1B is primarily involved in DNA damage response pathways, it is likely to be predominantly intracellular.
The experimental design should include:
Cell preparation: For intracellular proteins, fixation and permeabilization are required. Use a fixative compatible with your antibody (typically 4% paraformaldehyde) followed by a permeabilization agent appropriate for nuclear proteins .
Controls:
Antibody selection and validation:
Protocol optimization:
Titrate antibody concentration (starting with manufacturer recommendations)
Optimize fixation and permeabilization conditions
Adjust incubation times and temperatures
Remember that understanding your antibody's specifics is crucial - knowing whether it targets intracellular or extracellular epitopes determines your permeabilization approach .
Proper storage and handling of HUS1B antibodies are crucial for maintaining their activity and specificity. Based on manufacturer recommendations, the following conditions should be observed:
For Western blot applications using HUS1B antibodies, researchers should consider the following recommended dilutions and conditions:
Antibody dilution range: While specific HUS1B antibody dilution ranges weren't provided in the search results, comparable antibodies like HUS1 are typically used at dilutions of 1:300-1:1000 for Western blotting . As a starting point for HUS1B antibodies, a similar range can be tested, with optimization for your specific experimental conditions.
Sample preparation considerations:
Ensure complete protein denaturation if using a denaturing gel system
Include appropriate protease inhibitors in lysis buffers
Consider phosphatase inhibitors if studying phosphorylation status
Controls to include:
Positive control: Lysate from cells known to express HUS1B
Negative control: Lysate from cells with minimal HUS1B expression or HUS1B knockdown
Loading control: Housekeeping protein to ensure equal loading
Expected molecular weight: Be aware that HUS1B may be observed at a molecular weight close to the related HUS1 protein, which has a calculated molecular weight of 32 kDa and is observed at 32 kDa in Western blots .
Optimization tip: It is recommended that HUS1B antibodies should be titrated in each testing system to obtain optimal results, as the optimal dilution can be sample-dependent .
When detecting faint signals, researchers might consider using more sensitive detection systems or signal enhancement methods to improve visualization of low-abundance proteins like HUS1B.
Non-specific binding is a common issue when working with antibodies that can complicate data interpretation, especially with less well-characterized targets like HUS1B. Here are methodological approaches to identify and resolve non-specific binding:
Validation of observed bands/signals:
Optimization of blocking conditions:
Test different blocking agents (BSA, milk, commercial blockers)
Increase blocking time or concentration
Add 0.1-0.5% Tween-20 to reduce hydrophobic interactions
Antibody incubation adjustments:
Reduce primary antibody concentration
Optimize incubation temperature (4°C overnight vs. room temperature)
Add 0.1-0.5% Tween-20 to antibody dilution buffer
Consider adding low concentrations (0.1-0.5%) of non-homologous serum from the same species as your samples
Washing optimization:
Increase the number of washes
Extend washing duration
Use buffers with higher salt concentration or detergent content
Cross-reactivity assessment:
Test the antibody on samples known to lack HUS1B
Perform peptide competition assays to confirm specificity
Evaluate potential cross-reactivity with HUS1 and other related proteins
When analyzing results, carefully examine background levels and compare signal-to-noise ratios across different optimization conditions to determine the most specific detection parameters.
Differentiating between HUS1 and HUS1B signals is critical for accurate data interpretation, given their sequence similarities and related functions. Here's a methodological approach to ensure signal specificity:
Antibody selection strategy:
Choose antibodies raised against non-conserved regions between HUS1 and HUS1B
Verify antibody specificity through manufacturer validation data
Consider using antibodies that have been validated in knockout/knockdown systems
Experimental controls:
Include HUS1 knockout/knockdown samples to confirm HUS1B signal specificity
Similarly, include HUS1B knockout/knockdown samples to confirm HUS1 signal specificity
Use cell lines with differential expression of HUS1 versus HUS1B
Analytical approaches:
Functional discrimination:
When analyzing Western blot data, note that while both proteins have similar molecular weights, slight differences in migration patterns might be observable under high-resolution gel conditions. Always validate findings through multiple experimental approaches to confirm the identity of detected proteins.
When interpreting flow cytometry data for HUS1B detection, researchers should be aware of several common pitfalls that can lead to misinterpretation:
Autofluorescence interference:
Certain cell types exhibit high autofluorescence that can mask true signals
Always include unstained controls for each cell type
Consider using spectral compensation to correct for autofluorescence
Fixation and permeabilization artifacts:
Population gating challenges:
Inappropriate gating can exclude relevant cell populations or include debris
Use forward/side scatter profiles to exclude cell debris and doublets
Include viability dyes to exclude dead cells that often show non-specific binding
Signal intensity interpretation:
Low signal-to-noise ratio can make it difficult to distinguish positive from negative populations
Use isotype controls to establish background staining levels
Be cautious about interpreting small shifts in fluorescence as positive signals
Control-related issues:
Cell cycle effects:
As HUS1B is involved in cell cycle regulation, its expression may vary with cell cycle phase
Consider including cell cycle phase markers for more nuanced interpretation
Compare populations at similar cell cycle stages for more accurate comparisons
To improve data reliability, quantify signal-to-noise ratios, perform replicate experiments, and validate findings using complementary techniques such as Western blotting or immunofluorescence microscopy.
HUS1B antibodies can be powerful tools for investigating DNA damage response (DDR) pathways, given HUS1B's role in cell cycle checkpoint regulation. Here's a methodological approach for such studies:
Experimental design for DNA damage induction:
Use genotoxic agents (e.g., UV radiation, cisplatin, or hydroxyurea) to induce specific types of DNA damage
Apply time-course experiments to track HUS1B protein levels and localization following damage
Compare HUS1B responses to those of the better-characterized HUS1 protein
Co-immunoprecipitation studies:
Chromatin association studies:
Employ chromatin fractionation followed by Western blotting with HUS1B antibodies
Track HUS1B recruitment to chromatin following various types of DNA damage
Correlate chromatin association with cell cycle phases
Knockdown/knockout complementation:
In HUS1B-depleted cells, study the rescue effects of wild-type versus mutant HUS1B
Use HUS1B antibodies to confirm expression levels of exogenous proteins
Analyze checkpoint activation and cell survival outcomes
Comparative analysis with HUS1:
Given that HUS1B and HUS1 likely have distinct roles, use antibodies against both proteins to compare:
Temporal activation patterns
Subcellular localization changes
Protein stability following DNA damage
Interactions with other checkpoint proteins
Cell death pathway investigation:
When designing these experiments, ensure proper controls are in place, including both positive controls (known DDR proteins like γH2AX) and negative controls (non-DDR proteins) to validate your findings.
Studying protein phosphorylation is critical for understanding signaling cascades, particularly in DNA damage response pathways where HUS1B operates. Here's a methodological framework to investigate HUS1B phosphorylation:
Phosphorylation site prediction and analysis:
Utilize bioinformatics tools (NetPhos, PhosphoSitePlus) to predict potential phosphorylation sites on HUS1B
Compare conservation of these sites with the related HUS1 protein
Focus on kinase motifs relevant to DNA damage response (ATM/ATR, CHK1/CHK2)
Phosphorylation-specific detection methods:
Phospho-specific antibodies: If available, use antibodies recognizing specific phosphorylated residues
Phos-tag SDS-PAGE: Use this specialized gel system to separate phosphorylated from non-phosphorylated forms
Mass spectrometry: For unbiased identification of phosphorylation sites and stoichiometry
Phosphorylation induction and temporal dynamics:
Treat cells with DNA damaging agents and analyze HUS1B phosphorylation status over time
Use kinase inhibitors to identify responsible kinases
Compare phosphorylation patterns across different damage types (UV, IR, replication stress)
Functional significance assessment:
Generate phospho-mimetic (S/T→D/E) and phospho-deficient (S/T→A) mutants
Perform rescue experiments in HUS1B-depleted cells
Analyze effects on:
Protein-protein interactions (particularly with RAD1)
Chromatin association
Cell cycle checkpoint activation
Cell death induction
Comparative studies with HUS1:
Determine whether HUS1B shares phosphorylation sites with HUS1
Investigate whether the same kinases target both proteins
Analyze differences in phosphorylation-dependent functions
When conducting these experiments, researchers should be mindful that phosphorylation events may be transient and occur at low stoichiometry, requiring sensitive detection methods and careful optimization of experimental conditions.
Given HUS1B's involvement in cell cycle regulation and its ability to induce cell death when overexpressed, it represents a potential target for cancer research. Here's how HUS1B antibodies can be applied in oncology research contexts:
Expression profiling across cancer types:
Use HUS1B antibodies for immunohistochemistry (IHC) analysis of tissue microarrays
Compare HUS1B expression between normal and tumor tissues
Correlate expression levels with clinical outcomes and tumor characteristics
Consider using the same approach as validated for HUS1 antibodies, which have established IHC protocols
Functional studies in cancer models:
Investigate the consequences of HUS1B modulation (overexpression/knockdown) in cancer cell lines
Use HUS1B antibodies to confirm manipulation efficiency
Monitor effects on:
Cell proliferation
Apoptosis resistance
DNA damage response efficiency
Chemotherapy/radiotherapy sensitivity
Mechanism of HUS1B-induced cell death:
Synthetic lethality screening:
Identify drugs or genetic alterations that create synthetic lethality with HUS1B status
Use HUS1B antibodies to stratify cell lines or patient samples by expression level
Test whether HUS1B expression predicts sensitivity to DNA-damaging agents or checkpoint inhibitors
Biomarker potential assessment:
Evaluate HUS1B as a potential diagnostic or prognostic biomarker
Develop standardized protocols for HUS1B detection in clinical samples
Correlate HUS1B with established biomarkers of DNA damage response
Drug development applications:
Use HUS1B antibodies in target engagement studies for drug candidates
Screen for compounds that modulate HUS1B expression or function
Monitor HUS1B as a pharmacodynamic marker in treatment response
When conducting these studies, it's essential to validate findings across multiple cancer models and correlate experimental results with clinical data whenever possible to establish translational relevance.
Single-cell analysis technologies offer unprecedented insights into cellular heterogeneity and can be valuable for studying HUS1B in complex biological contexts:
Single-cell RNA sequencing (scRNA-seq) applications:
Profile HUS1B transcript levels across individual cells in heterogeneous populations
Correlate HUS1B expression with cell cycle phase markers
Identify cell subpopulations with distinctive HUS1B expression patterns
Analyze co-expression patterns with other DNA damage response genes
Single-cell protein analysis methods:
Mass cytometry (CyTOF): Incorporate HUS1B antibodies into CyTOF panels to simultaneously measure multiple proteins
Single-cell Western blotting: Quantify HUS1B protein levels in individual cells
Imaging flow cytometry: Combine flow cytometry with microscopy to correlate HUS1B levels with subcellular localization
Spatial transcriptomics and proteomics:
Visualize HUS1B expression patterns within tissue architecture
Correlate HUS1B distribution with microenvironmental features
Examine HUS1B expression in relation to tissue pathology
Integrative multi-omics approaches:
Combine single-cell transcriptomics, proteomics, and epigenomics
Create comprehensive maps of HUS1B regulation and function
Identify cell state-specific roles of HUS1B
Live-cell analysis of HUS1B dynamics:
Use antibody fragments or nanobodies for live-cell imaging
Track HUS1B protein dynamics in response to DNA damage
Monitor interactions with partners like RAD1 using proximity ligation techniques
When implementing these advanced approaches, researchers should be mindful of technical challenges, including antibody specificity validation in each new platform, potential fixation artifacts, and the need for computational expertise to analyze complex datasets.
Developing multiplexed assays that incorporate HUS1B detection allows researchers to study this protein in the context of broader signaling networks. Here are methodological considerations for successful multiplexing:
Antibody panel design principles:
Antibody clone selection: Choose HUS1B antibodies with minimal cross-reactivity
Host species diversification: Select antibodies from different host species to enable simultaneous detection
Fluorophore/reporter selection: Consider spectral overlap and signal strength when choosing reporters
Epitope accessibility: Ensure fixation and permeabilization conditions preserve all target epitopes
Validation requirements for multiplexed assays:
Single-stain controls: Test each antibody individually before combining
Fluorescence minus one (FMO) controls: Essential for setting accurate gates
Blocking optimization: Prevent non-specific binding in complex antibody mixtures
Signal-to-noise optimization: Adjust antibody concentrations to achieve balanced signal intensities
Platform-specific considerations:
Flow cytometry: Compensation and panel design to minimize spectral overlap
Mass cytometry: Metal-conjugated antibodies require validation for sensitivity and specificity
Multiplexed immunofluorescence: Sequential staining may be needed to avoid host species conflicts
Multiplexed Western blotting: Consider protein size to avoid signal overlap
Data analysis approaches:
Dimensionality reduction techniques (tSNE, UMAP) for visualizing high-parameter data
Clustering algorithms to identify cell populations with distinct patterns
Correlation analyses between HUS1B and other markers
Machine learning classification of cell states based on marker patterns
Biological context optimization:
When implementing multiplexed assays, thorough validation of each component and careful optimization of staining protocols are essential to ensure reliable, interpretable results.
The field of HUS1B research stands to benefit significantly from emerging technologies and methodological advances. Based on current knowledge gaps and technological trends, several promising research directions include:
Enhanced antibody development: Creation of more specific, sensitive antibodies against HUS1B, including phospho-specific antibodies to study post-translational modifications.
Application of proximity-based techniques: Implementation of proximity ligation assays and BioID approaches to better characterize HUS1B protein interactions in situ.
CRISPR-based functional genomics: Systematic analysis of HUS1B function through CRISPR knockout, knockin, and base editing technologies.
Single-cell and spatial biology approaches: Integration of HUS1B analysis in single-cell multi-omics and spatial transcriptomics/proteomics to understand context-dependent functions.
Translational research applications: Investigation of HUS1B as a potential biomarker or therapeutic target, particularly given its role in cell death induction and potential implications for cancer treatment resistance .