HSF1 Antibody

Heat Shock Transcription Factor-1, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to HSF1 Antibody

HSF1 (Heat Shock Factor 1) antibodies are immunological tools designed to detect and study the heat shock transcription factor 1, a master regulator of cellular stress responses and protein homeostasis. These antibodies enable precise visualization and quantification of HSF1 in diverse experimental settings, including Western blotting, immunohistochemistry (IHC), immunoprecipitation (IP), and chromatin immunoprecipitation (ChIP). HSF1 antibodies are critical for understanding its role in cancer biology, therapeutic resistance, and stress adaptation mechanisms .

Key Features of HSF1 Antibodies

Antibody TypeClone/SourceSpecies ReactivityApplications
Monoclonal (Rat IgG1)10H8Human, Mouse, RatWB, IP, IHC, IF
Rabbit Polyclonalab2923Human, Mouse, C. elegansWB, IP, ICC/IF
Rabbit Monoclonal (D3L8I)#12972Human, Mouse, Rat, Monkey, Bovine, Dog, PigWB, IP, IHC, ChIP
Mouse Monoclonal (IgG1)CD01/1B5HumanWB, IHC

Applications and Detection Methods

HSF1 antibodies are employed across multiple experimental platforms to elucidate HSF1’s subcellular localization, protein interactions, and transcriptional activity.

Western Blotting (WB)

Used to quantify HSF1 protein levels and confirm knockdown efficiency. For example, HSF1 antibodies detect a ~57–82 kDa band corresponding to the canonical protein .

Immunohistochemistry (IHC)

Identifies nuclear HSF1 accumulation in cancer tissues. High nuclear HSF1 levels correlate with aggressive tumor phenotypes in breast, ovarian, and hepatocellular cancers .

Chromatin Immunoprecipitation (ChIP)

Maps HSF1 binding to heat shock response element (HSE) regions in gene promoters. This method revealed HSF1’s role in regulating chaperones (e.g., HSP70, HSP90) and oncogenic pathways in malignant cells .

Immunofluorescence (IF)

Visualizes HSF1 translocation from cytoplasm to nucleus under stress conditions, such as heat shock or proteotoxic stress .

Cancer Prognosis and HSF1 Expression

High nuclear HSF1 levels are linked to poor outcomes in multiple cancers:

Cancer TypeHSF1 AssociationImpact on SurvivalSource
Breast (ER-positive)Increased nuclear HSF1HR = 2.10 (mortality risk)
Epithelial OvarianOverexpressionBiomarker for malignancy
HepatocellularElevated mRNA/proteinSensitivity to HSP90 inhibitors

HSF1’s prognostic value is independent of traditional markers like tumor stage or grade, making it a potential standalone biomarker .

Therapeutic Sensitization

HSF1 knockdown enhances cancer cell susceptibility to HSP90 inhibitors (e.g., NVP-HSP990):

Cell LineHSF1 KnockdownEffect on HSP90 Inhibitor IC50/LD50
A375 (Melanoma)shRNAIC50 ↓ 3.2×
Hep3B (HCC)shRNA + DoxycyclineLD50 ↓ 1.9×
HCT116 (Colorectal)shRNALD50 ↓ 6.5×

Targeting HSF1 in Cancer Therapy

HSF1 inhibition disrupts protein homeostasis and synergizes with HSP90 inhibitors to induce apoptosis. Preclinical models show tumor regression when combining HSF1 knockdown with HSP90 blockade .
Key Mechanisms:

  1. Chaperone Network Disruption: Reduces HSP70/HSP90 levels, impairing oncoprotein folding.

  2. Metabolic Stress Induction: Disrupts glucose metabolism and cell-cycle progression.

  3. Immune Modulation: Alters tumor microenvironment to enhance antitumor immunity .

Antibody Validation in Preclinical Studies

Antibodies like 10H8 and D3L8I validate HSF1 knockdown efficacy in:

  • Xenograft Models: HSF1 shRNA + NVP-HSP990 caused 76% tumor regression in melanoma .

  • Primary Cells: HSF1 suppression in myeloma cells triggered caspase-mediated death .

Table 2: Clinical Associations of HSF1 Expression

Cancer TypeHSF1 LocalizationPrognostic Impact
BreastNuclear accumulation↑ Mortality (HR = 1.62)
OvarianCytoplasmic/nuclearDiagnostic marker
HepatocellularElevated mRNASensitivity to HSP90i

Product Specs

Introduction
The transcription of heat-shock genes is rapidly induced after temperature stress in response to environmental changes, and it is also involved in oogenesis, spermatogenesis, and placental development. Hsp90 is a major repressor of the HSF1 gene. HSF1 is known for its participation in the heat shock response. The HSF1 protein regulates the transcription of hundreds of targets, such as genes involved in protein folding, detoxification, energy generation, carbohydrate metabolism, and cell wall organization. HSF1 binds to MTA1 in vitro & in breast carcinoma. Suppression of estrogen-dependent transcription may contribute to the role of HSF1 in cancer. Human cancer lines of diverse origins express higher dependence on HSF1 function to maintain proliferation and survival than their non-transformed counterparts. HSF1 induces anticoagulation and relaxation factors in vascular endothelial cells and could therefore be used to treat cardiovascular disease. HSF1 is directly involved in the regulation of HO1.
Formulation
1 mg/ml containing PBS, pH 7.4, 0.02% Sodium Azide, and 10% Glycerol.
Storage Procedures
For periods up to 1 month, store at 4°C; for longer periods, store at -20°C. Prevent freeze-thaw cycles.
Stability / Shelf Life
12 months at -20°C. 1 month at 4°C.
Applications
HSF1 antibody has been tested by ELISA and Western blot analysis to assure specificity and reactivity. Since application varies, however, each investigation should be titrated by the reagent to obtain optimal results. The recommended dilution range for Western blot analysis is 1:1,000–2,000. The recommended starting dilution is 1:1,000.
Synonyms
HSF-1, HSF1, HSTF-1, HSTF1, Heat shock factor protein 1, Heat shock transcription factor 1.
Purification Method
HSF1 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PJ7F9AT.
Immunogen
Anti-human HSF1 mAb, is derived from hybridization of mouse FO myeloma cells with spleen cells from BALB/c mice immunized with recombinant human HSF1 amino acids 1-529 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is HSF1 and what are its primary functions in cellular physiology?

HSF1 is a stress-inducible, DNA-binding transcription factor that orchestrates the heat shock response (HSR) in cells. It plays a central role in activating the transcription of heat shock proteins (HSPs), which function as molecular chaperones to protect cellular proteins from damage during stress conditions . HSF1 is widely expressed across diverse tissue types and can be found in both nuclear and cytoplasmic compartments, with its localization shifting in response to cellular stress . As the master regulator of the proteotoxic stress response, HSF1 remains in a monomeric inactive state under normal conditions but undergoes trimerization and nuclear translocation when activated by stressors like heat, oxidative damage, or chemical insults . This activation process involves multiple post-translational modifications that fine-tune HSF1's transcriptional activity. Beyond the classical heat shock response, HSF1 has been implicated in development, cancer progression, and aging-related processes, making it a multifaceted protein of significant research interest.

What structural features of HSF1 should researchers consider when selecting antibodies?

When selecting HSF1 antibodies, researchers should consider several key structural features that influence antibody recognition and experimental outcomes. HSF1 has distinct functional domains including a DNA-binding domain, trimerization domains (leucine zippers), a regulatory domain, and a transactivation domain . Up to two different isoforms have been reported for human HSF1, which may affect epitope accessibility depending on the antibody's target region . The protein undergoes various post-translational modifications (phosphorylation, sumoylation, acetylation) that can mask epitopes or alter protein conformation, potentially affecting antibody binding . The observed molecular weight of HSF1 often differs from its calculated mass of 57.3 kDa, typically appearing at 68-80 kDa in Western blots due to these modifications . When choosing antibodies, researchers should determine whether they need to detect specific phosphorylated forms (such as phospho-S326) which are indicators of HSF1 activation state . Additionally, since HSF1 is evolutionarily conserved with orthologs in multiple species (mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken), cross-reactivity considerations are important for comparative studies .

What are the optimal dilutions and conditions for different HSF1 antibody applications?

Determining optimal dilutions and conditions for HSF1 antibody applications requires careful consideration of experimental variables and antibody characteristics. For Western blotting (WB), HSF1 antibodies typically perform best at dilutions between 1:1000-1:4000, with optimization needed based on protein concentration and detection system sensitivity . When performing immunohistochemistry (IHC), starting dilutions of 1:500-1:2000 are recommended, with antigen retrieval preferably using TE buffer at pH 9.0, though citrate buffer at pH 6.0 may also be effective . For immunofluorescence (IF) or immunocytochemistry (ICC), dilutions of 1:200-1:800 typically yield optimal signal-to-noise ratios . Immunoprecipitation (IP) applications generally require 0.5-4.0 μg of antibody per 1.0-3.0 mg of total protein lysate to effectively capture HSF1 complexes . Buffer composition significantly impacts HSF1 detection—phosphatase inhibitors are essential when studying phosphorylated forms of HSF1, while protease inhibitor cocktails help prevent degradation during sample preparation. Temperature conditions during incubation can affect antibody binding kinetics and specificity, with overnight incubations at 4°C often producing cleaner results than shorter incubations at room temperature. The table below summarizes recommended starting parameters for different applications, though these should be optimized for each experimental system:

ApplicationRecommended DilutionOptimal IncubationSpecial Considerations
Western Blot1:1000-1:4000Overnight at 4°CVerify 68-80 kDa band
IHC1:500-1:20001-2 hours at RTTE buffer pH 9.0 retrieval
IF/ICC1:200-1:8001-2 hours at RTFixation method affects epitope accessibility
IP0.5-4.0 μg per 1-3 mg lysateOvernight at 4°CGentle washing to preserve complexes

How should researchers prepare samples for optimal HSF1 detection?

Sample preparation significantly impacts the successful detection of HSF1 using antibody-based methods. Cell lysis protocols require careful consideration as HSF1's subcellular localization changes between cytoplasm and nucleus depending on activation state . For comprehensive HSF1 detection, researchers should employ RIPA or other denaturing buffers containing both nuclear and cytoplasmic fractions. When studying HSF1 activation dynamics, separate nuclear and cytoplasmic fractions may provide more informative results. Protein extraction from tissues demands more rigorous homogenization methods while maintaining sample integrity, with flash-freezing of samples immediately after collection being critical to preserve HSF1's phosphorylation state . Phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate) are essential in all buffers when studying HSF1 activation, as phosphorylation at specific residues (particularly S326) serves as a key activation marker . Protease inhibitor cocktails prevent degradation during extraction, while reducing agents like DTT or β-mercaptoethanol help maintain proper protein conformation by preventing non-specific disulfide bond formation. Sample storage conditions significantly impact HSF1 integrity—aliquoting samples and storing at -80°C prevents freeze-thaw cycles that can degrade the protein. For immunohistochemistry applications, the fixation method substantially affects epitope accessibility, with formalin fixation times requiring optimization, and paraffin-embedded samples benefiting from antigen retrieval using TE buffer at pH 9.0 for optimal HSF1 detection .

What controls should be included when using HSF1 antibodies in experiments?

Including proper controls is essential for robust experimental design and valid interpretation of HSF1 antibody results. Primary controls should include both positive and negative tissue/cell controls—HEK-293, HeLa, A431, HepG2, K-562, and NIH/3T3 cells have been validated for positive HSF1 expression in Western blotting, while specific human tissues like lung cancer and testis tissue have been confirmed for immunohistochemistry . For antibody validation, tissues or cells from HSF1 knockout models serve as the gold standard negative control to confirm antibody specificity. Loading controls must be carefully selected based on experimental context—for whole cell lysates, housekeeping proteins like GAPDH or β-actin are appropriate, while nuclear fraction analysis should use nuclear-specific loading controls such as Lamin B or Histone H3 . Technical controls should include antibody-only controls (no primary antibody) to assess secondary antibody non-specific binding and isotype controls matching the primary antibody's host species and isotype to identify non-specific binding from the antibody class itself. Peptide competition assays provide powerful validation by demonstrating signal reduction when the antibody is pre-incubated with its target antigen peptide . Activation state controls are particularly important for HSF1 research—samples from heat-shocked cells (typically 42°C for 1 hour) should show increased nuclear localization and phosphorylation compared to non-stressed cells. For phospho-specific HSF1 antibodies, controls should include phosphatase-treated samples to confirm phospho-specificity of the detected signal .

Why does HSF1 sometimes appear at different molecular weights in Western blots?

The discrepancy between HSF1's calculated molecular weight (57.3 kDa) and its observed migration pattern (68-80 kDa) in Western blots results from several biological and technical factors . Post-translational modifications constitute the primary reason for the observed higher molecular weight. HSF1 undergoes extensive phosphorylation at multiple serine residues during activation, with phosphorylation at S326 being a particularly important marker of transcriptionally active HSF1 . These phosphorylation events can add significant apparent mass to the protein. Additionally, HSF1 can be subject to SUMOylation, acetylation, and other modifications that further alter its migration pattern in SDS-PAGE. The protein's intrinsic structural properties also influence migration—HSF1 contains regions with high negative charge density that can bind less SDS than typical proteins, resulting in reduced mobility during electrophoresis. Different experimental conditions can affect observed molecular weight; variations in gel percentage, buffer systems, and running conditions may shift the apparent molecular weight by several kilodaltons. When researchers observe unexpected banding patterns, they should consider several verification approaches: (1) using phosphatase treatment to eliminate phosphorylation-dependent shifts, (2) comparing heat-shocked versus non-shocked samples to observe activation-dependent mobility shifts, (3) implementing gradient gels to better resolve HSF1 isoforms, and (4) confirming identity through mass spectrometry or immunoprecipitation followed by Western blotting. The table below summarizes common HSF1 banding patterns and their likely interpretations:

Observed MWLikely InterpretationVerification Approach
57-60 kDaUnmodified/inactive HSF1Compare with heat-shocked samples
68-75 kDaPhosphorylated active HSF1Treat with phosphatase to see shift
75-80 kDaHyperphosphorylated/SUMOylated HSF1Use modification-specific antibodies
Multiple bandsMixture of different modification statesFractionate samples by cellular compartment

How can researchers overcome weak or non-specific HSF1 antibody signals?

Addressing weak or non-specific HSF1 antibody signals requires systematic troubleshooting of both experimental conditions and antibody selection. For weak signals, antibody concentration should first be optimized, potentially using higher concentrations (1:500 for Western blot instead of 1:2000) while monitoring background levels . Signal enhancement techniques include extended primary antibody incubation (overnight at 4°C rather than 1-2 hours at room temperature), using more sensitive detection systems like chemiluminescent substrates with longer activation periods, or employing signal amplification methods such as biotinylated secondary antibodies with streptavidin-HRP conjugates. Sample preparation critically impacts signal strength—increasing protein loading (50-100 μg for Western blots), improving extraction efficiency with more stringent lysis buffers, and enriching for nuclear fractions where activated HSF1 accumulates can all enhance detection . When facing non-specific binding, blocking conditions should be optimized by trying different blocking agents (5% non-fat dry milk versus 3-5% BSA) and extending blocking times to 2 hours at room temperature or overnight at 4°C. Washing protocols should be adjusted to include more washes with higher detergent concentrations (0.1-0.3% Tween-20) for more stringent conditions. If these approaches fail, switching antibody types may help—if a polyclonal antibody shows non-specific binding, trying a monoclonal targeting a different epitope might provide cleaner results . Cross-adsorbed secondary antibodies can reduce cross-reactivity in multi-species experiments. For particularly challenging samples, pre-adsorption of the primary antibody with cell/tissue lysates lacking HSF1 can remove antibodies that bind to non-specific epitopes.

What factors affect HSF1 antibody performance in different tissue types?

HSF1 antibody performance varies considerably across tissue types due to tissue-specific factors that influence epitope accessibility and background levels. Tissue fixation methods dramatically impact HSF1 detection—excessive formalin fixation can mask epitopes through protein cross-linking, while insufficient fixation may not adequately preserve tissue architecture . Optimization of antigen retrieval methods is particularly important for HSF1 detection in formalin-fixed, paraffin-embedded (FFPE) tissues, with heat-induced epitope retrieval using TE buffer at pH 9.0 generally yielding superior results compared to citrate buffer at pH 6.0 for many HSF1 antibodies . Tissue-specific HSF1 expression levels must be considered when interpreting results—HSF1 is widely expressed but at varying levels across tissues, with particularly high expression in testis tissue which serves as an excellent positive control . Endogenous peroxidase activity in tissues like liver, kidney, and certain tumor types can cause high background in IHC when using HRP-based detection systems; this requires effective quenching with hydrogen peroxide before antibody application. Tissues with high biotin content (liver, kidney, brain) may produce false-positive results when using biotin-streptavidin detection systems, necessitating biotin blocking steps or alternative detection methods . Background autofluorescence presents challenges in immunofluorescence applications, particularly in tissues with high collagen content or lipofuscin (common in aged tissues); this can be mitigated with Sudan Black B treatment or specialized quenching reagents. The microenvironment of certain tissues affects HSF1 detection—high-fat tissues require optimized deparaffinization steps, while tissues with abundant mucins may benefit from additional washing steps with higher detergent concentrations.

How can researchers effectively use HSF1 antibodies to study stress response mechanisms?

Investigating stress response mechanisms using HSF1 antibodies requires sophisticated experimental approaches that capture the dynamic nature of HSF1 activation and function. Stress kinetics studies should employ time-course experiments combining subcellular fractionation with Western blotting to track HSF1 translocation from cytoplasm to nucleus following stress exposure . Using phospho-specific antibodies that target key sites like S326 allows researchers to monitor HSF1 activation status, as phosphorylation at this residue correlates strongly with transcriptional activity . Chromatin immunoprecipitation (ChIP) assays using HSF1 antibodies enable mapping of HSF1 binding to heat shock elements (HSEs) in promoter regions of target genes, with several validated antibodies demonstrating high efficiency in ChIP applications . For visualizing dynamic HSF1 responses in living systems, immunofluorescence microscopy with HSF1 antibodies can track the formation of nuclear stress bodies—distinct subnuclear structures where HSF1 accumulates during stress . Co-immunoprecipitation using HSF1 antibodies allows identification of interacting partners that regulate HSF1 activity, with 0.5-4.0 μg of antibody per 1-3 mg of protein lysate typically yielding optimal results . Advanced imaging techniques like proximity ligation assays (PLA) can visualize HSF1 interactions with specific partners in situ, while super-resolution microscopy reveals the spatial organization of HSF1 within nuclear stress bodies at nanometer resolution. For high-throughput screening of HSF1 modulators, researchers can employ cell-based assays where HSF1 antibodies detect activation status following compound treatment, often in microscopy-based formats amenable to automation.

What strategies exist for investigating HSF1 post-translational modifications?

Investigating HSF1 post-translational modifications (PTMs) requires specialized antibodies and methodologies to capture these transient yet functionally critical states. Phospho-specific antibodies targeting key regulatory sites like S326, S303, and S121 provide direct visualization of HSF1 activation status . Western blotting with these antibodies should be performed using phosphatase inhibitor-supplemented buffers (sodium fluoride, sodium orthovanadate, and β-glycerophosphate) to preserve modification states. Phos-tag™ acrylamide gels offer enhanced resolution of phosphorylated HSF1 species compared to standard SDS-PAGE, allowing separation of multiply phosphorylated forms that would otherwise appear as a single broad band. For comprehensive PTM mapping, researchers can employ immunoprecipitation with general HSF1 antibodies followed by mass spectrometry analysis, though this approach requires significant protein amounts (typically immunoprecipitation from 5-10 mg of total protein) . 2D gel electrophoresis separating proteins first by isoelectric point then by molecular weight can resolve different HSF1 modified forms, with subsequent Western blotting using HSF1 antibodies to identify specific spots. Immunofluorescence microscopy using modification-specific antibodies can reveal the subcellular localization of modified HSF1 pools, particularly important given that certain modifications (like S303 phosphorylation) are associated with cytoplasmic retention while others (S326 phosphorylation) correlate with nuclear localization and activity . In vitro kinase assays using recombinant HSF1 as substrate followed by Western blotting with phospho-specific antibodies can identify kinases responsible for specific modifications. For studying dynamic modification patterns, pulse-chase experiments with metabolic labeling followed by HSF1 immunoprecipitation provide temporal resolution of modification sequences.

How can HSF1 antibodies be used in multiplexed detection systems?

Multiplexed detection systems incorporating HSF1 antibodies enable researchers to simultaneously visualize HSF1 alongside its regulatory partners or downstream targets. Multicolor immunofluorescence combines differentially labeled HSF1 antibodies with antibodies against interacting proteins or stress markers, requiring careful antibody selection to avoid cross-reactivity . Primary antibodies from different host species (rabbit anti-HSF1 with mouse anti-HSP70) minimize cross-reactivity issues. For Western blotting applications, multiplexed fluorescent detection systems using secondary antibodies conjugated to spectrally distinct fluorophores allow simultaneous visualization of HSF1 and other proteins without stripping and reprobing membranes. This approach is particularly valuable for monitoring HSF1 alongside its phosphorylated forms or downstream targets like HSP70 and HSP90. Mass cytometry (CyTOF) using metal-conjugated HSF1 antibodies enables high-dimensional analysis of HSF1 states in heterogeneous cell populations, though this requires specialized equipment. Imaging mass cytometry extends this approach to tissue sections, allowing spatial resolution of HSF1 activity patterns across different tissue regions. Sequential immunoprecipitation strategies can resolve HSF1 complexes—first immunoprecipitating with HSF1 antibodies, then using antibodies against suspected partners on the precipitated material. For highly multiplexed imaging, cyclic immunofluorescence methods involving iterative antibody staining, imaging, and signal removal allow visualization of dozens of targets on the same sample. When implementing these advanced approaches, validation experiments must confirm that antibody performance remains consistent in multiplexed formats, as steric hindrance between closely binding antibodies may affect signal intensity or specificity compared to single-target detection.

How should researchers interpret variability in HSF1 detection across different experimental systems?

Interpreting variability in HSF1 detection across experimental systems requires consideration of both biological and technical factors. Cell type-specific differences in HSF1 expression levels and activation states represent genuine biological variability—basal HSF1 levels differ across cell lines, with documented consistent detection in HEK-293, HeLa, A431, HepG2, K-562, and NIH/3T3 cells, while expression patterns in primary cells often differ from immortalized lines . Stress response thresholds vary between cell types, with some activating HSF1 at lower stress levels than others, creating apparent discrepancies in HSF1 activation when using standardized stress protocols. Cell culture conditions significantly impact HSF1 status—confluence levels, passage number, and serum conditions all affect basal HSF1 activation, potentially leading to contradictory results between laboratories. Technical variables include antibody lot-to-lot variability, particularly with polyclonal antibodies where epitope recognition can differ between batches . Detection system sensitivity influences apparent HSF1 levels, with more sensitive chemiluminescent systems potentially detecting HSF1 in samples where less sensitive colorimetric methods show negative results. To address these variability sources, researchers should implement standardized positive controls (like heat-shocked HeLa cells) across experiments, quantify relative rather than absolute HSF1 levels when comparing different systems, and use multiple antibodies recognizing different HSF1 epitopes to confirm detection patterns . Statistical analysis should account for this inherent variability—biological replicates should be derived from different passages or animals rather than technical replicates from the same source, and non-parametric statistical tests may be more appropriate than parametric tests when analyzing highly variable HSF1 data.

What are the best practices for quantifying HSF1 activation using antibody-based methods?

Accurate quantification of HSF1 activation using antibody-based methods requires careful attention to experimental design and analysis approaches. Western blot quantification should employ phospho-specific antibodies (particularly against S326) as the primary metric of HSF1 activation, with results normalized to total HSF1 levels rather than housekeeping genes to account for possible changes in total HSF1 expression . Densitometry analysis must include linear range validation for both total and phospho-HSF1 signals, as chemiluminescent detection can quickly saturate with highly expressed proteins. Nuclear-to-cytoplasmic ratio quantification via subcellular fractionation followed by Western blotting provides a powerful activation metric, with clean fractionation confirmed using compartment-specific markers (GAPDH for cytoplasm, Lamin B for nucleus) . In immunofluorescence approaches, quantification should include both intensity measurements and subcellular distribution analysis—automated image analysis tools can measure nuclear-to-cytoplasmic fluorescence intensity ratios across many cells, providing statistically robust activation metrics . Flow cytometry using HSF1 phospho-specific antibodies offers high-throughput quantification of activation in cell populations, though this requires effective permeabilization and fixation protocols to access intracellular HSF1. ChIP-qPCR quantifying HSF1 binding to heat shock elements in target gene promoters provides a functional measure of activation, though results must be normalized to input DNA and expressed relative to a non-HSF1 target region . For time-course experiments, area under the curve (AUC) analysis of activation metrics provides a comprehensive measure of the total HSF1 response. Whatever the approach, researchers should implement standardized positive controls (cells heat shocked at 42°C for 1 hour) and negative controls (non-stressed cells) in every experiment to enable normalization and comparison across experimental batches.

Product Science Overview

Introduction

Heat Shock Transcription Factor-1 (HSF1) is a crucial protein that plays a significant role in the cellular response to stress. It is highly conserved across eukaryotes and is the primary mediator of transcriptional responses to proteotoxic stress. This article delves into the background, structure, function, and significance of HSF1, particularly focusing on the mouse anti-human HSF1 antibody.

Structure of HSF1

HSF1 consists of several domains that regulate its binding and activity . The key domains include:

  1. DNA-Binding Domain (DBD): This N-terminal domain is the most conserved region in the HSF protein family. It recognizes the sequence nGAAn on target DNA, which constitutes heat shock elements (HSEs) for active HSF1 trimers to bind.
  2. Oligomerization Domain (Leucine Zipper Domains): These regions are responsible for oligomerization between HSF1 monomers. Under non-stress conditions, spontaneous HSF1 activation is negatively regulated by the interaction between these domains.
  3. Regulatory Domain (RD): Situated between the oligomerization domains, the RD regulates the Trans-Activation Domain (TAD) through negative control by repressing TAD in the absence of stress.
  4. Trans-Activation Domain (TAD): This C-terminal region contains two TADs (TAD1 and TAD2) that are crucial for HSF1’s transcriptional activation.
Function of HSF1

HSF1 functions as a stress-inducible and DNA-binding transcription factor that plays a central role in the transcriptional activation of the heat shock response (HSR). Upon exposure to heat and other stress stimuli, HSF1 undergoes homotrimerization and activates HSP gene transcription through binding to site-specific HSEs present in the promoter regions of HSP genes . This leads to the expression of a large class of molecular chaperones, heat shock proteins (HSPs), that protect cells from cellular insult damage.

In unstressed cells, HSF1 is present in a HSP90-containing multichaperone complex that maintains it in a non-DNA-binding inactivated monomeric form. Upon heat shock stress, HSF1 forms a chromatin-associated complex with TTC5/STRAP and p300/EP300 to stimulate HSR transcription, thereby increasing cell survival .

Mouse Anti-Human HSF1 Antibody

The mouse anti-human HSF1 antibody is an IgG1 κ monoclonal antibody that detects the HSF1 protein of mouse, rat, and human origin. It is used in various applications such as Western Blot (WB), Immunoprecipitation (IP), Immunofluorescence (IF), Immunohistochemistry (IHC), and Enzyme-Linked Immunosorbent Assay (ELISA) .

Significance of HSF1

HSF1 is involved in several critical cellular processes beyond the heat shock response. It plays a role in the regulation of lifespan, development, and metabolism. HSF1 also has important functions in non-stress regulation, such as activating transcription of transcription factor FOXR1, which in turn activates transcription of heat shock chaperones HSPA1A and HSPA6, and the antioxidant NADPH-dependent reductase DHRS2 .

Moreover, HSF1 has been implicated in various diseases, including cancer. Genetic elimination of HSF1 has been shown to protect mice from tumors induced by mutations in the RAS oncogene or a hot spot mutation in the tumor suppressor gene P53 . HSF1 promotes apoptotic cell death of pachytene spermatocytes exposed to thermal stress, indicating its role in quality control during spermatogenesis .

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.