The HSF5 antibody is a specialized monoclonal antibody designed to detect the Heat Shock Factor 5 (HSF5) protein, a transcription factor critical for male germ cell development and immune system modulation. HSF5 is a member of the heat shock factor family, traditionally associated with stress responses, but recent studies highlight its unique roles in spermatogenesis and tumor immunity. The antibody enables researchers to study HSF5 localization, expression, and functional interactions in cellular and tissue contexts .
Source: Hybridoma Technology
The HSF5 antibody (e.g., mAb10C3) was developed using hybridoma technology, which involves immunizing mice with testis antigens and screening hybridoma clones for specificity . Key findings include:
Specificity: mAb10C3 binds exclusively to HSF5 in early embryonic development (E7.5) and adult testis tissue, distinguishing it from other HSF family members .
Validation: Immunohistochemistry confirmed the antibody’s reactivity with spermatogonia and spermatocytes, validated against commercial anti-HSF5 antibodies (e.g., ab98939) .
Spermatogenesis: Studies using the antibody reveal HSF5’s essential role in meiotic prophase progression. HSF5 knockout mice exhibit arrested spermatogenesis at the pachytene stage, underscoring its requirement for chromatin organization and gene expression during male germ cell development .
Imaging: The antibody enables visualization of HSF5 in pachytene spermatocytes and round spermatids via immunostaining, with signal peaking during stages VII–VIII of spermatogenesis .
Lung Adenocarcinoma (LUAD): The antibody has been used to correlate HSF5 expression with immune infiltration and prognosis. High HSF5 levels are associated with increased tumor-infiltrating B cells (r = 0.439, P = 2.86e-24) and CD8+ T cells (r = 0.301, P = 1.21e-11), suggesting its role as a biomarker for immune modulation .
Immune Checkpoint Analysis: HSF5 expression correlates with ICOS (r = 0.62, P < 0.001) and BTLA (r = 0.58, P < 0.001), highlighting its potential in immunotherapy research .
Therapeutic Applications: Exploring HSF5’s role in tumor immunity could inform cancer immunotherapy strategies.
Reproductive Disorders: Investigating HSF5’s potential as a biomarker for male infertility.
Mechanistic Studies: Elucidating HSF5’s DNA-binding motifs and transcriptional targets under non-stress conditions .
HSF5 antibody may function as a transcriptional factor.
HSF5 (Heat Shock Factor Protein 5/Heat Shock Transcription Factor 5) is a member of the heat shock factor family with distinctive characteristics that set it apart from canonical HSFs. Unlike HSF1, HSF2, and HSF4 which possess two heptad repeats (HR-A and HR-B) that form inter-molecular leucine zippers for homotrimer oligomerization, HSF5 lacks these heptad repeats . HSF5 contains a winged-helix-turn-helix (WHTH)-like DNA-binding domain but recognizes DNA motifs different from typical heat shock elements recognized by other canonical HSFs . Most significantly, HSF5 functions under non-stress conditions rather than heat stress conditions, making it an atypical HSF .
HSF5 shows highly tissue-specific expression, predominantly in the testes. RT-PCR analysis demonstrates that HSF5 mRNA is specifically detected in juvenile and adult mouse testes but not in other adult organs . This contrasts with the ubiquitous expression pattern of HSF1, HSF2, and HSF4 . At the cellular level, immunostaining studies show that HSF5 protein appears in mid-pachytene spermatocyte nuclei (starting at stage VI seminiferous tubules) and continues to be expressed in spermatocyte nuclei through stages VII-XII and in round spermatids of stages I-VI . HSF5 is not detected in spermatogonia, pre-mid-pachytene spermatocytes, or elongated spermatids . Functionally, HSF5 is essential for meiotic prophase progression beyond the pachytene stage in male germ cells and is critical for male fertility .
HSF5 antibodies are valuable tools in reproductive biology and male fertility research. Common applications include:
These applications enable researchers to study HSF5 expression patterns, subcellular localization, and potential roles in normal and pathological conditions related to male reproduction .
When selecting an HSF5 antibody, researchers should consider:
Antibody type: Polyclonal antibodies offer broader epitope recognition but potentially more cross-reactivity, while monoclonal antibodies (e.g., mAb10C3) provide higher specificity for particular epitopes .
Species reactivity: Verify the antibody's reactivity with your species of interest. Available HSF5 antibodies show reactivity to various combinations of human, mouse, and rat HSF5 .
Validated applications: Confirm the antibody has been validated for your intended application (WB, IHC, IF, ELISA). For example, some HSF5 antibodies are validated for all applications, while others may be validated only for specific applications like WB and ELISA .
Target region: Consider whether the antibody targets specific domains of HSF5 that may be relevant to your research question. Some antibodies are raised against full-length HSF5, while others target specific peptide sequences .
Validation data: Review available validation data, including Western blot bands at the expected molecular weight (approximately 65 kDa for human HSF5) and specificity controls such as HSF5 knockout tissues .
Thorough validation of HSF5 antibodies should include:
Positive and negative tissue controls: Test the antibody on tissues with known HSF5 expression (testis) and tissues with minimal expression (e.g., liver, kidney) . Comparative analysis with a known anti-HSF5 antibody can serve as a reference point for validation .
Genetic controls: When available, use tissues from HSF5 knockout models as negative controls to confirm antibody specificity .
Peptide competition assays: Pre-incubate the antibody with the immunizing peptide to confirm binding specificity.
Multiple application validation: Verify antibody performance across multiple techniques (WB, IHC, IF) to ensure consistent results.
Cross-reactivity assessment: Test for potential cross-reactivity with other HSF family members, particularly in experimental systems where multiple HSF proteins may be expressed.
Titration experiments: Determine optimal working concentrations for each application by testing a range of antibody dilutions (e.g., 1:200-1:800 for IF or 1:500-1:3000 for WB) .
To maintain HSF5 antibody efficacy:
Store antibodies at -20°C for long-term storage, as recommended by manufacturers .
Aliquot antibodies before storage to avoid repeated freeze-thaw cycles that can degrade antibody quality . Some suppliers note that aliquoting may be unnecessary for -20°C storage in certain formulations .
Store in appropriate buffer conditions, typically PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 .
Antibodies are generally stable for one year after shipment when stored properly .
For working solutions, keep at 4°C for short-term use (typically up to one week).
Always follow manufacturer-specific recommendations, as formulations may vary between suppliers.
HSF5 antibodies can provide valuable insights into male infertility through several advanced approaches:
Comparative expression analysis: Use HSF5 antibodies to compare expression patterns in testicular biopsies from fertile individuals versus infertile patients. Research has shown that patients with azoospermia and low modified Johnson scores exhibit low expression of HSF5 .
Stage-specific spermatogenesis analysis: Since HSF5 expression begins in mid-pachytene spermatocytes (from stage VI seminiferous tubules) and continues through subsequent stages , use co-immunostaining with markers like SYCP3 (axial elements), γH2AX (DSBs and XY body), and H1t (mid-pachytene marker) to assess whether meiotic progression is affected in infertility models .
XY body association analysis: HSF5 signals show association with XY chromosomes in approximately 23% of pachytene spermatocytes . This association can be analyzed using confocal microscopy with whole-mount immunostaining to assess whether abnormalities in this pattern correlate with infertility phenotypes.
DNA damage response evaluation: In HSF5 knockout models, persistent γH2AX signals throughout nuclei and BRCA1 appearance along autosomes indicate unrepaired DNA damage . HSF5 antibodies can be used alongside DNA damage markers to assess whether similar defects occur in infertility models.
Chromatin organization assessment: Since HSF5 binds to promoters of genes associated with chromatin organization , immunoprecipitation techniques using HSF5 antibodies can help identify chromatin abnormalities in infertility cases.
Researchers may encounter several challenges when working with HSF5 antibodies:
Weak signal intensity: HSF5's restricted expression pattern may result in weak signals. To address this:
Optimize antibody concentration through careful titration
Extend primary antibody incubation time (overnight at 4°C)
Use signal amplification systems like tyramide signal amplification
Consider antigen retrieval methods for fixed tissues
Background staining: Especially in testicular tissue which may have high background. Solutions include:
Increase blocking time and concentration (5% BSA or normal serum)
Add 0.1-0.3% Triton X-100 to reduce non-specific binding
Use more stringent washing steps (0.1% Tween-20 in PBS)
Consider testing multiple antibodies from different sources
Cross-reactivity with other HSF family members: HSF family proteins share some sequence homology. To minimize this:
Select antibodies raised against unique regions of HSF5
Validate with negative controls from HSF5 knockout tissues
Perform Western blot to confirm specificity based on molecular weight
Stage-dependent expression variations: Since HSF5 expression is stage-dependent in seminiferous tubules , consistent staging of tubules is essential for comparative analysis. Use stage-specific markers (SYCP3, γH2AX, STRA8, H1t) for accurate staging.
Epitope masking due to protein interactions: HSF5's association with chromatin may mask epitopes. Try multiple antibodies targeting different regions of HSF5, or consider non-denaturing conditions for certain applications.
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) with HSF5 antibodies can reveal genome-wide binding patterns of HSF5. Consider these critical aspects:
Antibody selection: Choose antibodies specifically validated for immunoprecipitation. Antibodies that recognize the DNA-binding domain may be less effective if this domain is involved in DNA interactions.
Crosslinking optimization: Since HSF5 is a transcription factor, standard formaldehyde crosslinking (1% for 10 minutes) may be appropriate, but optimization may be required.
Cell type selection: Use cell populations enriched for HSF5 expression, such as isolated pachytene spermatocytes, to improve signal-to-noise ratio.
Control experiments: Include:
Input chromatin as a normalization control
IgG control to assess non-specific binding
Ideally, chromatin from HSF5 knockout tissues as a negative control
Validation of binding sites: Since HSF5 recognizes DNA motifs different from typical heat shock elements , validate identified binding sites using:
ChIP-qPCR for selected targets
Motif analysis to identify HSF5-specific binding sequences
Reporter assays to confirm functional significance
Data analysis focus: Analyze binding patterns with particular attention to:
Monoclonal and polyclonal HSF5 antibodies offer distinct advantages and limitations:
Research has shown that monoclonal antibodies like mAb10C3 are highly specific for HSF5 detection in embryonic development and adult testis tissue sections . This antibody has been validated through comparison with commercial anti-HSF5 antibodies (e.g., ab98939) and demonstrates specific detection of HSF5 in spermatogonia and spermatocytes .
Polyclonal antibodies offer broader epitope recognition and are available from multiple commercial sources, with demonstrated reactivity in human, mouse, and rat samples .
The reliability of detection methods varies by experimental context:
Protein expression levels (Western blot):
Tissue localization (Immunohistochemistry/Immunofluorescence):
Developmental studies:
Protein-DNA interactions (ChIP):
Single-cell analysis:
Immunofluorescence of isolated germ cells provides higher resolution of HSF5 localization
Flow cytometry can be used for quantitative analysis of HSF5-positive cell populations
Essential for distinguishing stage-specific expression patterns
Distinguishing genuine HSF5 signals from artifacts requires rigorous controls and methodological considerations:
Essential controls:
Genetic controls: Use HSF5 knockout tissues as definitive negative controls
Absorption controls: Pre-incubate antibody with immunizing peptide to confirm signal specificity
Secondary antibody controls: Omit primary antibody to assess non-specific secondary antibody binding
Isotype controls: Use matched isotype IgG at the same concentration as the primary antibody
Validation through multiple antibodies:
Expected HSF5 localization patterns:
Nuclear localization in mid-pachytene spermatocytes through round spermatids
Absence from spermatogonia, pre-mid-pachytene spermatocytes, and elongated spermatids
Association with XY chromosomes in approximately 23% of pachytene spermatocytes
Signal predominantly on chromatin loops rather than on axes in pachytene spermatocytes
Signal confounders to consider:
Autofluorescence: Common in testicular tissue; use specific filters or spectral unmixing
Cross-reactivity with other HSF family members: Verify molecular weight by Western blot
Fixation artifacts: Compare different fixation methods (paraformaldehyde, methanol)
Stage-dependent expression: Ensure proper staging of seminiferous tubules using markers
Quantitative approach:
Establish signal-to-noise ratio thresholds based on control samples
Use digital image analysis with consistent thresholding parameters
Quantify signal intensity across multiple samples and biological replicates
Research has established important correlations between HSF5 expression and male fertility disorders:
Azoospermia: Patients with azoospermia and low modified Johnson scores show reduced expression of HSF5 , suggesting a potential diagnostic role for HSF5 immunostaining in male infertility evaluation.
Meiotic arrest: HSF5 knockout mice exhibit male-specific infertility characterized by spermatocyte arrest at the pachytene stage . Similar arrest patterns may be identifiable in human infertility cases through HSF5 and stage-specific marker analysis.
DNA damage accumulation: HSF5 deficiency leads to persistence of γH2AX signals throughout spermatocyte nuclei and BRCA1 appearance along autosomes, indicating unrepaired DNA damage . This pattern may be observed in certain types of male infertility.
Meiotic sex chromosome inactivation (MSCI) failure: Disruption of HSF5 leads to failure of MSCI , a critical process for male fertility. MSCI failure has been associated with male infertility in multiple genetic conditions.
Sperm morphology abnormalities: HSF5 mutant males show reduced sperm count, increased sperm head size, and abnormal tail architecture , suggesting HSF5's role in sperm morphogenesis.
These correlations highlight the potential value of HSF5 antibodies in diagnostic and research applications for male infertility.
Multiple complementary approaches can elucidate HSF5's role in spermatogenesis:
Stage-specific expression analysis:
Genetic manipulation models:
Protein-DNA interaction studies:
Proteomic approaches:
Co-immunoprecipitation with HSF5 antibodies to identify protein interaction partners
Mass spectrometry to identify post-translational modifications of HSF5
Proximity labeling techniques (BioID, APEX) to identify proximity interactors
Developmental timing analysis:
HSF5 antibodies can serve as valuable tools in evaluating therapeutic approaches for HSF5-related infertility:
Therapeutic target validation:
Use HSF5 antibodies to confirm target engagement of small molecules or biologics designed to modulate HSF5 activity
Assess changes in HSF5 expression, localization, or DNA binding in response to treatment
Phenotypic screening:
Screen potential therapeutic compounds for their ability to rescue HSF5 expression or localization in cellular models of HSF5 dysfunction
Validate hits using multiple HSF5 antibodies to confirm specificity
Biomarker development:
Use HSF5 immunostaining as a biomarker to stratify infertile patients who might benefit from specific therapeutic approaches
Correlate HSF5 expression patterns with treatment responsiveness
Gene therapy assessment:
Evaluate HSF5 re-expression following gene therapy approaches in HSF5-deficient models
Monitor restoration of normal spermatogenesis using HSF5 and stage-specific markers
Monitoring treatment effects in animal models:
Track changes in HSF5 expression, localization, and downstream effects following experimental treatments
Correlate molecular changes with fertility restoration
Assess sperm parameters and testicular histology alongside HSF5 immunostaining
Developmental timing interventions:
By incorporating HSF5 antibodies into therapeutic evaluation pipelines, researchers can gain mechanistic insights into treatment effects and develop more targeted approaches for HSF5-related male infertility.
Several critical questions about HSF5 biology remain unresolved and could be addressed using antibody-based approaches:
HSF5 post-translational modifications: What post-translational modifications regulate HSF5 activity? Phospho-specific or other modification-specific HSF5 antibodies could identify regulatory modifications and their timing during spermatogenesis.
Transcriptional mechanisms: How does HSF5 regulate gene expression during meiotic prophase? ChIP-seq studies with HSF5 antibodies combined with transcriptome analysis could identify direct target genes and regulatory mechanisms.
Protein interaction network: What proteins interact with HSF5 to regulate meiotic progression? Co-immunoprecipitation studies with HSF5 antibodies followed by mass spectrometry could reveal the HSF5 interactome.
Chromatin loop association: What is the functional significance of HSF5's association with chromatin loops rather than axes ? High-resolution imaging with HSF5 antibodies combined with chromatin conformation capture techniques could elucidate this relationship.
Species-specific differences: Are there species-specific differences in HSF5 function? Comparative immunostaining studies across species could reveal evolutionary conservation and divergence in HSF5 biology.
XY body association mechanism: What is the mechanism and significance of HSF5's association with XY chromosomes in a subset of pachytene spermatocytes ? Super-resolution microscopy with HSF5 antibodies could provide insights.
Developmental transitions: What signals trigger HSF5 expression in mid-pachytene spermatocytes? Temporal analysis of HSF5 expression in relation to other developmental signals could address this question.
Integration of HSF5 antibodies with cutting-edge technologies offers exciting opportunities:
Spatial transcriptomics with protein detection:
Combine HSF5 immunostaining with spatial transcriptomics (e.g., Visium, MERFISH) to correlate HSF5 protein localization with gene expression patterns in intact testicular tissue
Identify spatially restricted gene expression programs that depend on HSF5 activity
Single-cell multi-omics:
Integrate HSF5 antibody-based cell sorting with single-cell RNA-seq and ATAC-seq to correlate HSF5 expression with transcriptional and chromatin accessibility changes
Identify cell state transitions associated with HSF5 expression
In situ protein interaction detection:
Apply proximity ligation assays (PLA) with HSF5 antibodies to visualize and quantify protein interactions in intact tissue
Identify spatially and temporally restricted interactions during spermatogenesis
Live cell imaging:
Develop cell-permeable HSF5 antibody fragments or nanobodies for live imaging of HSF5 dynamics during meiosis in cultured spermatocytes
Track real-time changes in HSF5 localization during critical transitions
Organoid models:
Use HSF5 antibodies to validate and characterize testicular organoid models for studying spermatogenesis in vitro
Monitor HSF5 expression as a marker of proper meiotic progression in organoid systems
CRISPR screening with HSF5 readouts:
Develop high-content screening approaches using HSF5 antibodies as readouts for CRISPR screens targeting regulators of meiotic progression
Identify genetic factors that modulate HSF5 expression or function
Several methodological innovations could enhance the utility of HSF5 antibodies:
Domain-specific antibodies:
Development of antibodies targeting specific functional domains of HSF5 (DNA-binding domain, transcriptional regulatory domains)
Creation of conformation-specific antibodies that recognize active versus inactive HSF5 states
Improved signal amplification methods:
Application of tyramide signal amplification or other signal enhancement techniques to detect low-abundance HSF5 in specific cell types
Development of branched DNA amplification methods for simultaneous detection of HSF5 protein and mRNA
Multiplex imaging platforms:
Adaptation of HSF5 antibodies for multiplex immunofluorescence techniques (e.g., Vectra, CODEX) to simultaneously visualize HSF5 alongside multiple markers
Integration with multiplexed ion beam imaging (MIBI) or imaging mass cytometry for highly multiplexed protein detection
Reversible immunolabeling methods:
Development of cleavable linker systems for HSF5 antibodies to allow sequential immunostaining rounds on the same specimen
Application to create comprehensive maps of protein expression in relation to HSF5
High-throughput validation platforms:
Creation of testicular tissue microarrays covering all stages of spermatogenesis for rapid validation of new HSF5 antibodies
Development of cell line models with controlled HSF5 expression for antibody validation
Antibody engineering approaches:
Generation of bispecific antibodies that simultaneously recognize HSF5 and key interacting partners
Development of antibody fragments with improved tissue penetration for whole-mount applications