HSF4 Antibody

Shipped with Ice Packs
In Stock

Description

Lens Development and Cataract Studies

HSF4 antibodies have been pivotal in elucidating HSF4’s role in lens fiber cell differentiation. Key findings include:

  • HSF4-null mice exhibit cataracts due to defective organelle degradation and γ-crystallin downregulation .

  • HSF4 regulates autophagy via ATG9a activation, a mechanism validated using HSF4 antibodies in knockout models .

Cancer Research

In renal cell carcinoma (RCC), HSF4 antibodies revealed its oncogenic role:

  • HSF4 knockdown reduced RCC cell proliferation and invasion by suppressing MET signaling .

  • HSF4 overexpression correlates with poor prognosis, making it a therapeutic target .

Stress Response and DNA Repair

HSF4 antibodies identified its interaction with COIL under UV exposure, stabilizing R-loops to modulate inflammation and senescence genes .

Western Blot Protocols

  • Dilution: 1:1,000–1:4,000 (Proteintech) .

  • Buffer: Tris-glycine SDS-PAGE, transferred to PVDF membranes .

Functional Assays

  • Chromatin Immunoprecipitation (ChIP): HSF4 antibodies confirmed direct binding to γ-crystallin and FGF promoters in lens cells .

  • Immunofluorescence: Nuclear HSF4 localization post-UV exposure was visualized in NIH-3T3 cells .

Product Stability

  • Storage: -20°C long-term; avoid freeze-thaw cycles .

  • Formats: Liquid (PBS with 0.02% sodium azide) .

Species Cross-Reactivity

  • Most antibodies target human and mouse HSF4, with limited reactivity in zebrafish or rat .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery times.
Synonyms
Cataract; Marner antibody; CTM antibody; Heat shock factor protein 4 antibody; Heat shock transcription factor 4 antibody; hHSF4 antibody; HSF 4 antibody; HSF4 antibody; HSF4_HUMAN antibody; HSTF 4 antibody
Target Names
HSF4
Uniprot No.

Target Background

Function
HSF4 is a DNA-binding protein that specifically binds heat shock promoter elements (HSE). It functions as a transcriptional repressor and activator.
Gene References Into Functions
  1. A novel mutation in HSF4 was identified in a large British pedigree, causing dominant congenital lamellar cataract. PMID: 29243736
  2. High HSF4 expression is an independent indicator of poor overall survival and recurrence-free survival in patients with primary colorectal cancer. PMID: 29131521
  3. In cultured human lens epithelial cells, HSF4 stabilizes and retains p53 in the nucleus to activate target genes such as fas cell surface death receptor (Fas) and Bcl-2-associated X apoptosis regulator (Bax). PMID: 28981088
  4. A novel homozygous HSF4 mutation (c.521T>C, p.Leu174Pro) was reported in two siblings with congenital cataracts. PMID: 26490182
  5. BCAS2 interacts with HSF4 and negatively regulates its protein stability via ubiquitination. PMID: 26319152
  6. HSF4 might act as a switch between lens epithelial cell proliferation and secondary fiber cell differentiation, a process primarily dependent on p53. PMID: 25940838
  7. It was concluded that the new mutation of c.331C>T in HSF4 DNA may be responsible for the autosomal dominant congenital cataract in this family. PMID: 25877371
  8. HSF4 p.Arg116His recreates the childhood lamellar cataract in mice, suggesting that incomplete penetrance associated with early cataracts may not be an absence but a limitation of phenotype detection. PMID: 24975927
  9. This is the first report of the novel missense mutation, c.69G-->T (p.K23N), in exon 3 of the HSF4 locus on 16q21-q22 associated with bilateral congenital cataracts in a Chinese family. PMID: 24637349
  10. Transcriptional activation of HSF4 is mediated by interactions between activator and repressor domains within the C-terminal end. PMID: 24045990
  11. HSF4 exerts its function on lens differentiation via positive regulation of DLAD expression. PMID: 23507146
  12. HSF4 and WRN CNVs might be involved in ARC pathogenesis in the Han Chinese. PMID: 23329665
  13. This provides the first evidence demonstrating that HSF4 plays a role in DNA damage repair and may contribute to a better understanding of congenital cataract formation. PMID: 22587838
  14. This study reports the absence of mutations in all studied genes in four families with phenotypes associating cataract, mental retardation, and microcephaly. PMID: 22103961
  15. HSF2 and HSF4 regulate transcription of HIF-1a and a critical balance between these HSFs is required to maintain HIF-a expression in a repressed state. PMID: 21258402
  16. Two missense mutations associated with age-related cataract did not, or only slightly, alter HSF4 activity, implying that other genetic and environmental factors affect the functions of these mutant proteins. PMID: 20670914
  17. Hsf4b could interact with and be phosphorylated by MAP kinase P38. PMID: 20564821
  18. A mutant DNA-binding domain of HSF4 is associated with autosomal dominant lamellar and Marner cataract. This suggests that HSF4 is crucial to lens development. PMID: 12089525
  19. This is the first report describing the association of an autosomal recessive cataract with the HSF4 locus on 16q21-q22.1 and the first description of HSF4 splice variants. PMID: 15277496
  20. HSF4 binds to alphaB-crystallin, Hsp70, and Hsp82 promoters and has a role in interacting with the canonical heat shock element of the alphaB-crystallin gene. PMID: 15308659
  21. Findings confirm that mutations in HSF4 may result in both autosomal dominant and autosomal recessive congenital cataract, and highlight the locus heterogeneity in autosomal recessive congenital cataract. PMID: 15959809
  22. The results indicate that in the absence of Hsf1 and Hsf2, Hsf4b expression in cells leads to an increased ability of Hsf4b to bind HSE during G1, resulting in enhanced synthesis of inducible Hsp70. PMID: 16552721
  23. These results identified a novel missense mutation R74H in the transcription factor gene HSF4 in a Chinese cataract family and expand the spectrum of HSF4 mutations causing cataract. PMID: 16876512
  24. This result indicates that HSF4 mutations account for only a small fraction of age-related cataracts. PMID: 18941546
  25. This study has shown the first nonsense mutation in HSF4 causing autosomal recessive cataracts in a large consanguineous family from Pakistan. PMID: 19014451

Show More

Hide All

Database Links

HGNC: 5227

OMIM: 116800

KEGG: hsa:3299

STRING: 9606.ENSP00000264009

UniGene: Hs.512156

Involvement In Disease
Cataract 5, multiple types (CTRCT5)
Protein Families
HSF family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in heart, skeletal muscle, eye and brain, and at much lower levels in some other tissues.

Q&A

What is HSF4 and why is it important in biomedical research?

HSF4 (heat shock factor 4) is a 53 kDa transcription factor belonging to the HSF family that specifically binds heat shock promoter elements (HSE). It exists in two primary isoforms: HSF4B, which activates transcription, and HSF4A, which represses transcription. HSF4 is critically important in lens development and maintenance, playing essential roles in denucleation and organelle degradation during lens fiber cell differentiation to maintain transparency . Research interest in HSF4 stems from its documented role in congenital cataracts, with multiple mutations identified in families with autosomal recessive cataract conditions . Additionally, HSF4 regulates expression of γ-crystallin and growth factors essential for cell growth and differentiation, making it a significant focus for developmental biology and ophthalmology research .

What applications can HSF4 antibodies be reliably used for?

Based on validated experimental protocols, HSF4 antibodies have been successfully employed in multiple applications including:

  • Western blotting (WB): Reliable detection of HSF4 protein at approximately 53 kDa under reducing conditions

  • Immunofluorescence/Immunocytochemistry: For visualizing cellular and subcellular localization of HSF4, particularly in nuclear regions

  • Electrophoretic Mobility Shift Assay (EMSA): For analyzing DNA-binding capabilities of HSF4 to heat shock elements

  • Flow cytometry: For quantifying HSF4 expression in different cell populations

  • ELISA: For quantitative detection of HSF4 protein

  • Chromatin Immunoprecipitation (ChIP): For examining in vivo binding of HSF4 to target gene promoters

What cell and tissue types show significant HSF4 expression?

While HSF4 mRNA is expressed ubiquitously at low levels, protein expression levels vary significantly across tissues. The lens shows exceptionally high HSF4 expression and activity compared to other tissues, where HSF4 constitutes the major HSE-binding activity . Other tissues with documented HSF4 protein expression include:

  • Brain and lung (at moderate levels)

  • Cell lines including NIH-3T3 mouse embryonic fibroblasts

  • Human LNCaP prostate cancer cell line

  • HeLa human cervical epithelial carcinoma cell line

Western blot analysis confirms HSF4 detection in these cell lines at approximately 53 kDa, making them suitable experimental models for HSF4 research .

How should researchers design experiments to differentiate between HSF4A and HSF4B isoforms?

Differentiation between HSF4 isoforms requires careful experimental design addressing their structural and functional differences:

Methodology for isoform differentiation:

  • RT-PCR with isoform-specific primers targeting the 45 amino acid region (aa 246-320) where HSF4A shows substitution compared to HSF4B .

  • Western blotting with antibodies recognizing shared epitopes, followed by precise molecular weight determination:

    • HSF4B: 493 amino acids (full length)

    • HSF4A: Shows distinctive migration pattern due to the 45 aa substitution

  • Functional assays:

    • Transcriptional reporter assays: HSF4B activates transcription while HSF4A represses it

    • DNA binding assays (EMSA): Both bind HSE elements but with potentially different affinities

  • Domain-specific antibodies targeted to the region of difference (aa 246-320) can provide definitive identification when available .

For comprehensive characterization, researchers should employ multiple approaches in parallel, as relying on a single method may lead to ambiguous results due to the high similarity between isoforms.

What controls should be included when validating HSF4 antibody specificity?

Rigorous validation of HSF4 antibody specificity requires multiple complementary controls:

Essential positive controls:

  • Recombinant HSF4 protein (such as E. coli-derived recombinant human HSF4 Ser309-Pro463)

  • Lysates from cells known to express HSF4 (lens tissue, NIH-3T3, LNCaP, or HeLa cells)

Essential negative controls:

  • HSF4-null tissue/cells (e.g., from HSF4 knockout mice)

  • Cells with confirmed absence of HSF4 expression

  • Pre-absorption with recombinant HSF4 antigen to demonstrate signal elimination

Additional validation approaches:

  • siRNA knockdown of HSF4 to confirm signal reduction

  • Competing antibody assays with multiple HSF4 antibodies recognizing different epitopes

  • Peptide competition assays using the specific immunogen peptide

  • Cross-reactivity assessment with other HSF family members (HSF1, HSF2) to confirm specificity

These combined approaches provide comprehensive evidence for antibody specificity and help avoid misinterpretation of experimental results.

How can researchers effectively study HSF4's DNA binding activity?

For comprehensive analysis of HSF4's DNA binding properties, researchers should implement the following methodological approach:

EMSA (Electrophoretic Mobility Shift Assay) protocol optimization:

  • Prepare nuclear extracts from lens tissue or cells expressing HSF4

  • Use labeled oligonucleotides containing validated HSE consensus sequences

  • Include competition controls with unlabeled HSE oligos

  • Include super-shift assays with HSF4 antibodies to confirm specificity

  • Compare with HSF1 binding patterns to identify HSF4-specific signatures

ChIP (Chromatin Immunoprecipitation) analysis:

  • Focus on genomic regions containing HSE consensus sequences

  • Target known HSF4-regulated genes (crystallins, FGF genes)

  • Use appropriate cross-linking conditions optimized for transcription factors

  • Include input controls and negative controls (IgG or non-related antibody)

  • Consider sequential ChIP (re-ChIP) to analyze co-binding with HSF1 or other factors

Reporter assays with HSE elements:

  • Design luciferase constructs with wild-type and mutated HSE elements

  • Compare activation patterns between HSF4A and HSF4B isoforms

  • Analyze the functional consequences of HSF4 mutations on DNA binding

These complementary approaches provide a comprehensive assessment of HSF4's DNA binding properties and transcriptional regulatory functions.

How can researchers effectively analyze HSF4 mutations identified in cataract patients?

To comprehensively analyze HSF4 mutations identified in cataract patients, researchers should implement a systematic multidisciplinary approach:

Structural and functional characterization:

  • Express recombinant wild-type and mutant HSF4 proteins (such as G199EfsX15, R405X, and M419GfsX29) with appropriate tags (e.g., FLAG-tag)

  • Conduct protein stability assays:

    • Cycloheximide chase experiments to assess protein turnover rates

    • Western blotting at multiple time points to quantify degradation kinetics

  • Evaluate subcellular localization:

    • Immunofluorescence microscopy to confirm nuclear trafficking

    • Subcellular fractionation with Western blotting for quantitative analysis

  • Assess DNA binding capabilities:

    • EMSA to measure HSE-mediated DNA binding efficiency

    • Quantify differences between wild-type and mutant proteins

  • Measure transcriptional activity:

    • Luciferase reporter assays with HSE-containing promoters

    • Analyze activation/repression patterns compared to wild-type

Domain-specific functional analysis:

  • For mutations affecting the DNA binding domain (e.g., A19D, R73H, I86V, L114P, R119C): Focus on DNA binding assays

  • For mutations affecting the trimerization domain (e.g., R175P): Evaluate oligomerization capacity

  • For C-terminal mutations: Analyze activation and repression domain functions

This comprehensive approach allows researchers to determine the specific molecular mechanisms by which different HSF4 mutations lead to cataract formation.

What methodologies can effectively distinguish between HSF1 and HSF4 activities in lens development research?

Distinguishing between HSF1 and HSF4 activities in lens development requires sophisticated approaches targeting their unique and overlapping functions:

Comparative binding and expression analysis:

  • Gel shift assays with lens extracts:

    • Use specific antibodies to super-shift HSF1 and HSF4 complexes

    • Quantify relative contribution of each factor to total HSE binding activity

  • Western blot analysis:

    • Compare expression levels of HSF1 and HSF4 in lens versus other tissues

    • Track developmental changes in HSF1:HSF4 ratios during lens maturation

Genetic approaches:

  • Single and double knockout models:

    • Compare HSF4-null, HSF1-null, and double-null phenotypes

    • Analyze restoration of normal phenotypes in double-null versus single knockouts

  • Target gene expression analysis:

    • Measure expression of heat shock proteins (Hsp27, Hsp60, Hsp70, Hsp90)

    • Analyze FGF expression patterns (FGF-1, FGF-4, FGF-7)

    • Compare γ-crystallin expression across genotypes

Competition analysis:

  • ChIP-seq to identify genomic regions where:

    • HSF1 and HSF4 bind exclusively

    • HSF1 and HSF4 compete for binding

    • HSF1 and HSF4 bind cooperatively

  • Sequential ChIP to directly assess co-occupancy of binding sites

These approaches collectively reveal the complex interplay between HSF1 and HSF4 in lens development, particularly how HSF1 competes with HSF4 for expression of FGFs and heat shock proteins.

How can researchers analyze the role of HSF4 in denucleation and organelle degradation during lens fiber differentiation?

Investigating HSF4's role in denucleation and organelle degradation during lens fiber differentiation requires specialized techniques:

Cellular and tissue-level analysis:

  • Histological and immunohistochemical approaches:

    • TUNEL assays to detect DNA fragmentation

    • Electron microscopy to assess organelle integrity

    • Immunostaining for nuclear envelope components

    • Tracking markers of lysosomal/autophagosomal activity

  • Live cell imaging with appropriate lens cell models:

    • Time-lapse microscopy of fluorescently labeled organelles

    • Analysis of denucleation kinetics with nuclear markers

Molecular mechanisms:

  • Analysis of HSF4 regulation of key genes:

    • ChIP analysis of HSF4 binding to DNASE2B promoter

    • Quantification of DNASE2B expression in wild-type versus HSF4-null lens

    • Assessment of α-crystallin B chain/CRYAB expression and its impact on lysosomal acidification

  • p53/TP53 regulation:

    • Analysis of p53 protein levels in lens fiber cells

    • Investigation of HSF4-mediated p53 stabilization mechanisms

    • Evaluation of p53-dependent pathways in organelle degradation

Experimental models:

  • Primary lens epithelial cell differentiation systems

  • Lens-specific HSF4 conditional knockout models

  • Transgenic models expressing mutant HSF4 forms

These multifaceted approaches allow researchers to dissect the complex role of HSF4 in the specialized process of lens fiber cell organelle elimination, which is critical for lens transparency.

How should researchers interpret contradictory HSF4 antibody results across different experimental systems?

When confronted with contradictory HSF4 antibody results across experimental systems, researchers should implement a systematic troubleshooting approach:

Source of variation analysis:

  • Antibody-related factors:

    • Epitope location: Different antibodies may recognize distinct domains of HSF4

    • Clone specificity: Compare monoclonal (e.g., clone #504524 or 2E7) versus polyclonal antibodies

    • Isoform recognition: Verify whether antibodies distinguish between HSF4A and HSF4B isoforms

  • Sample preparation variations:

    • Extraction methods: Nuclear versus whole-cell extracts may yield different results

    • Reducing conditions: HSF4 detection is optimal under specific reducing conditions

    • Buffer composition: Immunoblot Buffer Group 2 has been validated for HSF4 detection

  • Expression level variations:

    • Create a reference table comparing HSF4 expression across tissues and cell lines

    • Establish detection thresholds for each antibody

    • Normalize signals to appropriate loading controls

Resolution strategies:

  • Multi-antibody validation:

    • Test multiple antibodies recognizing different HSF4 epitopes

    • Compare results between different antibody clones and manufacturers

  • Correlation analysis:

    • Correlate protein detection with mRNA expression data

    • Validate with orthogonal methods (e.g., mass spectrometry)

  • Calibration curves:

    • Establish standard curves using recombinant HSF4 protein

    • Determine linear detection ranges for each antibody and system

This systematic approach helps identify the sources of variability and establish reliable protocols for consistent HSF4 detection across experimental systems.

What are the optimal storage and handling conditions for maintaining HSF4 antibody functionality?

To maintain optimal HSF4 antibody functionality and prevent experimental inconsistencies, researchers should adhere to these evidence-based guidelines:

Storage conditions and stability:

Storage PhaseTemperatureDurationSpecial Considerations
As supplied-20°C to -70°C12 monthsAvoid repeated freeze-thaw cycles
After reconstitution2°C to 8°C1 monthMaintain sterile conditions
Long-term after reconstitution-20°C to -70°C6 monthsStore in working aliquots

Critical handling protocols:

  • Use a manual defrost freezer to avoid temperature fluctuations

  • Prepare small working aliquots immediately after reconstitution

  • Clearly label aliquots with date of reconstitution and number of freeze-thaw cycles

  • Include carrier proteins (e.g., BSA) when diluting to working concentrations

Reconstitution guidelines:

  • Follow manufacturer-specific reconstitution protocols

  • Allow antibody to reach room temperature before opening

  • Reconstitute using sterile techniques

  • Mix gently by inversion rather than vortexing

Quality control measures:

  • Periodically test antibody functionality using positive controls

  • Include internal standards across experiments to track potential degradation

  • Document lot-to-lot variations when using new antibody batches

Adherence to these guidelines ensures consistent antibody performance across experiments and maximizes the useful lifespan of HSF4 antibodies.

How can researchers resolve non-specific binding issues when working with HSF4 antibodies?

When encountering non-specific binding with HSF4 antibodies, researchers should implement this systematic optimization protocol:

Blocking optimization:

  • Test multiple blocking agents:

    • BSA (1-5%)

    • Non-fat dry milk (1-5%)

    • Normal serum (from species other than antibody source)

    • Commercial blocking buffers

  • Optimize blocking time (1-16 hours) and temperature (4°C, RT)

Antibody dilution optimization:

  • Perform serial dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000)

  • Determine optimal signal-to-noise ratio for each application

  • For Western blotting, 2 μg/mL has been validated for Human/Mouse HSF4 Monoclonal Antibody

Buffer and washing protocol refinement:

  • Increase washing stringency (more washes, longer duration)

  • Adjust salt concentration in wash buffers (150-500 mM NaCl)

  • Add low concentrations of detergents (0.05-0.1% Tween-20)

  • For HSF4 Western blots, Immunoblot Buffer Group 2 has been validated

Cross-reactivity reduction:

  • Pre-absorb antibody with proteins from species or tissues showing cross-reactivity

  • Use monoclonal antibodies (e.g., clone #504524 or 2E7) for higher specificity

  • Consider using F(ab) fragments instead of whole IgG molecules

  • Validate with HSF4-null samples as definitive negative controls

Additional approach for persistent problems:

  • Peptide competition assays using specific epitope peptides

  • Immunoprecipitation followed by mass spectrometry to identify cross-reacting proteins

  • Epitope mapping to identify regions prone to cross-reactivity

Implementation of this protocol systematically eliminates sources of non-specific binding, resulting in cleaner, more interpretable experimental results with HSF4 antibodies.

How can researchers effectively integrate HSF4 antibody data with transcriptomic analyses?

To achieve meaningful integration of HSF4 antibody data with transcriptomic analyses, researchers should implement this methodological framework:

Experimental design for multi-omics integration:

  • Matched sampling:

    • Collect protein and RNA from the same biological samples

    • Process in parallel workflows to minimize technical variation

    • Include time-course analyses to capture dynamic relationships

  • ChIP-seq and RNA-seq correlation:

    • Perform HSF4 ChIP-seq to identify genome-wide binding sites

    • Correlate with RNA-seq data to identify direct transcriptional targets

    • Focus on HSF4-bound promoters showing expression changes in HSF4-null models

Analytical approaches:

  • Target gene validation workflow:

    • Identify HSF4-bound genes from ChIP-seq

    • Filter for genes with expression changes in transcriptomics

    • Validate with RT-qPCR in wild-type vs. HSF4-null or HSF4-mutant systems

    • Confirm protein-level changes with Western blotting

  • Pathway and network analysis:

    • Identify enriched pathways from integrated datasets

    • Compare HSF4-regulated gene networks with HSF1-regulated networks

    • Focus on lens-specific pathways involving γ-crystallins and FGFs

Implementation for known HSF4 targets:

Gene/ProteinAntibody Validation MethodTranscriptomic ValidationIntegration Approach
γ-crystallinsWestern blot, IHCRT-qPCR, RNA-seqDirect correlation analysis
FGF-1, FGF-4, FGF-7Western blot, IHCIn situ hybridization, RNA-seqInverse correlation analysis
Hsp27, Hsp60, Hsp70Western blot, IHCRT-qPCRTissue-specific correlation
DNASE2BChIP, Western blotRNA-seqDirect target analysis

This integrated approach provides comprehensive understanding of HSF4's regulatory network across molecular levels and identifies the most reliable biomarkers of HSF4 activity.

What methodological approaches can address contradictory findings between HSF4 antibody studies and genetic knockout models?

When confronted with contradictions between antibody-based studies and genetic models, researchers should implement this reconciliation framework:

Systematic contradiction analysis:

  • Characterize the specific nature of contradictions:

    • Target protein expression discrepancies

    • Subcellular localization differences

    • Binding partner inconsistencies

    • Functional outcome disparities

  • Evaluate knockout model integrity:

    • Confirm complete absence of HSF4 protein in knockouts

    • Assess potential compensatory mechanisms (especially HSF1 upregulation)

    • Evaluate potential artifacts from targeting strategy

  • Assess antibody reliability:

    • Validate antibody specificity in knockout tissues

    • Evaluate epitope accessibility in different contexts

    • Consider post-translational modifications affecting epitope recognition

Reconciliation strategies:

  • Tissue and developmental context analysis:

    • Compare results across different tissues (lens vs. non-lens)

    • Evaluate developmental stage-specific effects

    • Consider cell-type specific regulation

  • Isoform-specific analysis:

    • Determine if contradictions relate to specific HSF4 isoforms

    • Generate isoform-specific knockout models

    • Use isoform-discriminating antibodies when available

  • Molecular compensation analysis:

    • Examine HSF1-HSF4 competitive interactions

    • Compare single knockout to double knockout phenotypes

    • Analyze expression patterns of shared target genes

Example reconciliation approach for HSF4 regulation of heat shock proteins:

  • Contradictory finding: HSF4-null mice show increased expression of Hsp60, Hsp70, Hsp90 in lens epithelial cells despite HSF4 being a transcriptional activator

  • Resolution: Analysis of HSF1-HSF4 double-null mice revealed that HSF1 competes with HSF4 for regulation of these genes, explaining the paradoxical increase in HSF4-null mice

This systematic approach helps resolve contradictions between different experimental approaches and reveals complex regulatory mechanisms that might be missed by any single experimental methodology.

How can researchers effectively investigate HSF4's role in DNA repair through RAD51 regulation?

To comprehensively investigate HSF4's emerging role in DNA repair through RAD51 regulation, researchers should implement this multifaceted approach:

Molecular interaction analysis:

  • ChIP and promoter analysis:

    • Perform HSF4 ChIP followed by qPCR for RAD51 promoter regions

    • Identify specific HSE elements in the RAD51 promoter

    • Conduct site-directed mutagenesis of HSE elements to confirm functionality

  • Transcriptional regulation:

    • Compare RAD51 mRNA levels in wild-type versus HSF4-deficient cells

    • Perform luciferase reporter assays with RAD51 promoter constructs

    • Analyze HSF4 isoform-specific effects on RAD51 transcription

Functional DNA repair assessment:

  • Homologous recombination assays:

    • Use DR-GFP reporter systems in HSF4-expressing versus HSF4-knockdown cells

    • Quantify HR efficiency after inducing double-strand breaks

    • Rescue experiments with RAD51 overexpression in HSF4-deficient cells

  • DNA damage response analysis:

    • Monitor γH2AX foci formation and resolution

    • Track RAD51 foci formation after DNA damage

    • Analyze cell survival after genotoxic stress in relation to HSF4 status

Cell type-specific considerations:

  • Lens-specific analysis:

    • Compare DNA repair capacities in lens epithelial cells versus lens fiber cells

    • Correlate with developmental HSF4 expression patterns

    • Investigate potential connections to lens transparency maintenance

  • Cancer cell analysis:

    • Evaluate HSF4-RAD51 relationship in cancer cells (LNCaP, HeLa)

    • Assess implications for therapeutic resistance

    • Investigate potential synthetic lethality approaches

This comprehensive approach will elucidate the significance of HSF4-mediated RAD51 regulation in different cellular contexts and may reveal novel therapeutic targets for conditions involving DNA repair deficiencies.

What techniques can be used to investigate the interaction between HSF4 and p53/TP53 in lens fiber cells?

To investigate the emerging interaction between HSF4 and p53/TP53 in lens fiber cells, researchers should employ these specialized techniques:

Protein interaction analysis:

  • Co-immunoprecipitation strategies:

    • Perform reciprocal IPs (HSF4→p53 and p53→HSF4)

    • Use lens fiber cell extracts for physiological relevance

    • Compare interactions in stressed versus unstressed conditions

    • Include appropriate negative controls (IgG, HSF4-null samples)

  • Proximity ligation assay (PLA):

    • Visualize HSF4-p53 interactions in situ

    • Quantify interaction frequency in different lens regions

    • Compare wild-type versus mutant HSF4 proteins

Functional regulation assessment:

  • p53 stability analysis:

    • Cycloheximide chase experiments comparing p53 half-life

    • Ubiquitination assays in presence/absence of HSF4

    • Analysis of p53 post-translational modifications affected by HSF4

  • Transcriptional regulation:

    • ChIP-seq to identify shared and exclusive genomic targets

    • Reporter assays for p53-responsive elements with/without HSF4

    • RT-qPCR analysis of p53 target genes in HSF4-null lens

In vivo relevance:

  • Genetic interaction studies:

    • Compare phenotypes of HSF4-null, p53-null, and double-null lens

    • Analyze lens fiber cell apoptosis and differentiation

    • Assess lens transparency and cataract formation

  • Developmental timing analysis:

    • Track HSF4-p53 interactions throughout lens development

    • Correlate with critical stages of fiber cell differentiation

    • Analyze in relation to denucleation processes

Experimental model considerations:

  • Use primary lens epithelial cells undergoing differentiation

  • Employ lens-specific conditional knockout models

  • Consider transgenic models expressing mutant forms of HSF4 or p53

This comprehensive approach will elucidate the mechanistic details of HSF4-p53 interactions and their functional significance in lens development and maintenance.

What optimizations improve the detection sensitivity of HSF4 in tissues with low expression levels?

To enhance HSF4 detection in tissues with low expression levels, researchers should implement these evidence-based technical optimizations:

Sample preparation optimization:

  • Subcellular fractionation:

    • Focus on nuclear extracts where HSF4 is concentrated

    • Reduce cytoplasmic protein background

    • Optimize nuclear extraction buffers for transcription factor recovery

  • Protein concentration techniques:

    • TCA precipitation for dilute samples

    • Immunoprecipitation before Western blotting

    • Sequential extraction protocols to enrich for DNA-bound fraction

Western blotting enhancements:

  • Signal amplification strategies:

    • Use high-sensitivity chemiluminescent substrates

    • Implement biotin-streptavidin amplification systems

    • Consider tyramide signal amplification protocols

  • Optimized membrane and transfer conditions:

    • PVDF membrane validated for HSF4 detection

    • Reducing conditions critical for optimal detection

    • Immunoblot Buffer Group 2 specifically validated for HSF4

Sensitivity comparison across detection methods:

MethodRelative SensitivityOptimal ApplicationSpecial Considerations
Standard Western blotBaselineGeneral expression analysis2 μg/mL antibody concentration optimal
Chemiluminescent WB2-5× baselineLow expression tissuesExtended exposure times without background
IP-Western10-50× baselineVery low expressionAccount for IP efficiency
ImmunofluorescenceVariableLocalization studiesSignal amplification may be required
Flow cytometry2-10× baselineCell population analysisCareful permeabilization required

Validated protocol for low expression tissues:

  • Extract nuclear proteins using high-salt extraction

  • Concentrate samples using TCA precipitation

  • Resolve on 10% SDS-PAGE gels under reducing conditions

  • Transfer to PVDF-plus membrane

  • Block with optimized blocking buffer

  • Probe with 2 μg/mL HSF4 antibody (clone #504524 or 2E7)

  • Use high-sensitivity detection system with optimized exposure times

Implementation of these optimizations can significantly improve detection of HSF4 in tissues with naturally low expression levels, enabling more comprehensive analysis of HSF4 biology across diverse tissue types.

What considerations are important for multiplexing HSF4 with other heat shock factor antibodies in co-localization studies?

For successful multiplexing of HSF4 with other heat shock factor antibodies in co-localization studies, researchers should address these critical considerations:

Antibody selection and validation:

  • Cross-reactivity assessment:

    • Test each antibody individually against all HSF family members

    • Validate with knockout or knockdown controls for each HSF

    • Confirm specificity using peptide competition assays

  • Species compatibility analysis:

    • Select primary antibodies from different host species

    • Validate secondary antibody specificity against each primary

    • Test for cross-reactivity between secondary antibodies

Fluorophore selection strategies:

  • Spectral separation optimization:

    • Choose fluorophores with minimal spectral overlap

    • Perform single-color controls for spectral bleed-through

    • Consider linear unmixing for closely overlapping signals

  • Signal intensity balancing:

    • Match detection sensitivities across channels

    • Adjust antibody concentrations to achieve comparable signals

    • Consider the relative abundance of each target (HSF4 vs HSF1/HSF2)

Sample preparation considerations:

  • Epitope accessibility:

    • Optimize fixation protocols (duration, fixative type)

    • Evaluate different antigen retrieval methods for each antibody

    • Test different permeabilization conditions

  • Sequential versus simultaneous staining:

    • Compare results from both approaches

    • Consider sequential staining if antibodies compete for closely located epitopes

    • Document order effects if using sequential approach

Optimized multiplexing protocol for HSF1/HSF4 co-localization:

  • Fix samples in 4% PFA (10 minutes) followed by methanol permeabilization

  • Block with 5% normal donkey serum

  • Apply mouse anti-HSF4 (clone #504524 or 2E7) and rabbit anti-HSF1 antibodies

  • Detect with spectrally distinct secondary antibodies

  • Include DAPI nuclear counterstain

  • Image with confocal microscopy using sequential scanning

  • Perform quantitative colocalization analysis (Pearson's coefficient, Manders' overlap)

This optimized approach enables reliable visualization of HSF4 in relation to other heat shock factors, critical for understanding their functional interplay in various cellular contexts.

How can HSF4 antibodies be used to assess cataract development risk in clinical research?

For utilizing HSF4 antibodies in clinical research to assess cataract development risk, researchers should implement this translational methodology:

Biomarker development strategy:

  • Lens epithelial cell analysis from surgical specimens:

    • Collect anterior lens capsule specimens during cataract surgery

    • Process for both protein (Western blot/IHC) and RNA analysis

    • Compare HSF4 levels and localization with clinical parameters

  • Mutation-specific antibody applications:

    • Develop antibodies recognizing common HSF4 mutations

    • Validate in cell models expressing mutant HSF4 proteins

    • Evaluate specificity against wild-type HSF4

Clinical correlation approaches:

  • HSF4 target protein assessment:

    • Measure γ-crystallin levels as HSF4 activity biomarkers

    • Quantify FGF expression patterns in lens epithelial cells

    • Correlate with cataract severity and morphology

  • Risk stratification protocol:

    • Establish baseline HSF4 expression in normal lens epithelium

    • Define expression thresholds associated with cataract risk

    • Integrate with genetic screening for HSF4 mutations

Assay standardization for clinical application:

ParameterStandardization ApproachQuality Control Measure
Sample collectionStandardized surgical techniqueTissue area normalization
Protein extractionValidated protocol for small samplesSpiked-in standards
Antibody selectionValidated clone (#504524 or 2E7)Lot-to-lot validation
QuantificationDigital image analysisInternal calibration curves
Data normalizationHousekeeping protein controlsMulti-site validation

Potential clinical applications:

  • Family screening protocol for inherited cataracts:

    • Genetic testing for HSF4 mutations

    • Protein expression analysis where accessible

    • Integration with clinical examination findings

  • Age-related cataract risk assessment:

    • Evaluation of HSF4 activity in age-matched controls

    • Correlation with environmental risk factors

    • Development of predictive algorithms integrating HSF4 status

This translational approach bridges basic HSF4 antibody research with clinical applications, potentially enabling early risk assessment and intervention strategies for cataract development.

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.