HSF1 Human

Heat Shock Transcription Factor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Heat Shock Response (HSR)

HSF1 orchestrates HSR by:

  1. Sensing Stress: Directly detecting temperature changes via unfolding of the regulatory domain .

  2. Trimerization: Forming a homotrimer that binds HSEs in promoters of HSP genes (e.g., HSP70, HSP90) .

  3. Transcriptional Activation: Recruiting coactivators to drive cytoprotective gene expression .

Non-Stress Roles

HSF1 regulates:

  • Metabolism: Glucose utilization, lipid synthesis, and mitochondrial function .

  • Cellular Homeostasis: Protein folding, autophagy (via SQSTM1), and DNA repair .

  • Development: Embryonic growth and differentiation .

Cancer

HSF1 is constitutively active in tumors, promoting survival, chemoresistance, and metastasis. Key findings:

Cancer TypeHSF1 RoleClinical Correlation
HepatoblastomaDrives dedifferentiation via YAP1/β-catenin pathways Higher HSF1 levels correlate with tumor aggressiveness and mortality
Breast CancerPredicts poor prognosis, especially in ER-positive subtypes Nuclear HSF1 linked to increased mortality (HR: 2.10)
Pancreatic CancerSuppresses apoptosis by downregulating SMAC Associated with therapeutic resistance

HSF1 also upregulates glycolysis enzymes (e.g., LDHA, PDK3) and autophagy genes (e.g., ATG7), enhancing tumor growth and chemotherapy evasion .

Neuroprotection

HSF1 safeguards neurons from protein misfolding by:

  • HSP-Independent Mechanisms: Trimerization-deficient HSF1 mutants retain neuroprotective effects .

  • Gene Regulation: Upregulating 1,211 genes in healthy neurons, including non-HSP neuroprotective factors .

HSF1Base: Target Gene Database

The HSF1Base database catalogs 24,635 HSF1-regulated genes, including:

  • General Targets: HSP70, HSP90, DNAJB1 .

  • Tissue-Specific Targets: VDUP1 (breast cancer), HO1 (antioxidant response) .

  • Aging-Related Targets: SQSTM1, ULK1 (autophagy) .

Genetic Variants

Polymorphisms in HSF1 influence protein levels and function:

  • 3′UTR Variants: Modify microRNA binding, altering translational suppression (e.g., rs78202224) .

  • Coding SNPs: Predicted to disrupt structural integrity (e.g., C1263A) .

Therapeutic Targeting

HSF1 inhibition is explored for cancer treatment:

  • Mechanisms: Disrupt HSF1 trimerization or DNA binding .

  • Challenges: Avoiding toxicity to normal HSR-dependent tissues .

  • Biomarker Potential: HSF1 levels may predict response to HSP90 inhibitors .

Product Specs

Introduction
The heat-shock gene transcription process is rapidly activated following temperature stress as a response to environmental shifts. This process is also involved in oogenesis, spermatogenesis, and placental development. Hsp90, a significant repressor of the HSF1 gene, plays a role in this response. The HSF1 protein is known for its involvement in the heat shock response and regulates the transcription of numerous targets, including genes associated with protein folding, detoxification, energy production, carbohydrate metabolism, and cell wall organization. HSF1 has been observed to bind to MTA1 both in vitro and in breast carcinoma, suggesting that the suppression of estrogen-dependent transcription might contribute to HSF1's role in cancer development. Human cancer cell lines from various origins display a greater reliance on HSF1 function for sustained proliferation and survival compared to their non-transformed counterparts. Furthermore, HSF1 induces anticoagulation and relaxation factors within vascular endothelial cells, indicating its potential application in cardiovascular disease treatment. Additionally, HSF1 directly regulates HO1, contributing to its antioxidative function. Notably, both NF-kappaB and HSF1 are systemically activated in cases of human acute pancreatitis, and HSF1 activation has been shown to offer protection against the severity of pancreatitis. HSF acts as a critical transcription factor involved in the up-regulation of VDUP1 expression in response to stresses such as high cell density and serum deprivation in cultures.
Description
Recombinant Human HSF1, produced in E.Coli, is a single, non-glycosylated polypeptide chain composed of 549 amino acids (1-529). It possesses a molecular mass of 59.4kDa. This HSF1 protein is fused to a 20 aa His-Tag at its N-terminus.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The HSF1 protein is provided at a concentration of 1mg/ml in a buffer consisting of 20mM Tris pH 8, 50mM NaCl, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 75% as assessed by SDS-PAGE.
Synonyms
HSF-1, HSF1, HSTF-1, HSTF1, Heat shock factor protein 1, Heat shock transcription factor 1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MDLPVGPGAA GPSNVPAFLT KLWTLVSDPD TDALICWSPS GNSFHVFDQG QFAKEVLPKY FKHNNMASFV RQLNMYGFRK VVHIEQGGLV KPERDDTEFQ HPCFLRGQEQ LLENIKRKVT SVSTLKSEDI KIRQDSVTKL LTDVQLMKGK QECMDSKLLA MKHENEALWR EVASLRQKHA QQQKVVNKLI QFLISLVQSN RILGVKRKIP LMLNDSGSAH SMPKYSRQFS LEHVHGSGPY SAPSPAYSSS SLYAPDAVAS SGPIISDITE LAPASPMASP GGSIDERPLS SSPLVRVKEE PPSPPQSPRV EEASPGRPSS VDTLLSPTAL IDSILRESEP APASVTALTD ARGHTDTEGR PPSPPPTSTP EKCLSVACLD KNELSDHLDA MDSNLDNLQT MLSSHGFSVD TSALLDLFSP SVTVPDMSLP DLDSSLASIQ ELLSPQEPPR PPEAENSSPD SGKQLVHYTA QPLFLLDPGS VDTGSNDLPV LFELGEGSYF SEGDGFAEDP TISLLTGSEP PKAKDPTVS.

Q&A

Advanced Research Questions

  • What is the role of HSF1 in cancer progression and how can researchers systematically study its cancer-specific functions?

    HSF1 plays a significant role in cancer progression through mechanisms that extend beyond its classical stress response function. In cancer, HSF1:

    • Facilitates oncogenic transformation and maintenance of malignant phenotypes

    • Shows elevated nuclear levels across a wide range of cancers, correlating with poor survival

    • Influences genes related to various cellular processes beyond HSP induction

    • Acts as a critical dependency factor in multiple cancer types, with genetic elimination protecting mice from tumors induced by RAS oncogene or P53 mutations

    Methodologically, researchers can systematically study HSF1's cancer functions through:

    1. Clinical correlation studies:

      • Tissue microarrays and immunohistochemistry to assess HSF1 levels and localization in patient samples

      • Correlation of HSF1 expression with clinical parameters and survival outcomes using Kaplan-Meier analysis and Cox proportional hazard models

      • Analysis of public expression profiling data to correlate HSF1 mRNA levels with cancer-specific mortality

    2. Functional studies:

      • Genetic manipulation of HSF1 levels in cancer models using RNA interference or CRISPR-Cas9

      • Pharmacological inhibition using HSF1-specific inhibitors like I HSF115

      • Comparison of HSF1-dependent transcriptional programs in normal vs. cancer cells

    3. Mechanistic studies:

      • ChIP-seq to identify cancer-specific HSF1 binding sites

      • Proteomic analysis to identify cancer-specific HSF1 interactors

      • Metabolic profiling to understand HSF1's impact on cancer metabolism

    A notable study involving 1,841 participants in the Nurses' Health Study found that nuclear HSF1 levels were elevated in ~80% of in situ and invasive breast carcinomas. In invasive carcinomas, HSF1 expression was associated with high histologic grade, larger tumor size, and nodal involvement at diagnosis (P < 0.0001). Multivariate analysis showed that high HSF1 levels were independently associated with increased mortality (hazards ratio: 1.62; 95% CI: 1.21–2.17; P < 0.0013), particularly in estrogen receptor (ER)-positive breast cancers .

  • How can researchers develop and evaluate novel inhibitors targeting HSF1?

    Developing HSF1 inhibitors represents an important research direction given HSF1's role in cancer and other diseases. A systematic approach includes:

    1. Target identification and validation:

      • Structure-based design using available structural information on HSF1 domains (e.g., DNA-binding domain)

      • Identification of druggable pockets through computational modeling

      • Pharmacophore definition based on potential interactions with residues lining putative binding pockets

    2. Screening and hit identification:

      • Virtual screening of compound libraries using defined pharmacophores

      • Creation of biased sublibraries based on virtual screening results

      • Development of discriminating cell-based assays to identify compounds that specifically affect HSF1 activity

    3. Hit validation and development:

      • Direct binding assays (e.g., Surface Plasmon Resonance) to confirm interaction with HSF1 or its domains

      • Structure-activity relationship (SAR) studies to improve potency and selectivity

      • Assessment of compound effects on:

        • HSF1 oligomerization

        • Nuclear localization

        • DNA binding

        • Transcriptional activity

    4. Biological characterization:

      • Transcriptome analysis to assess compound effects on HSF1-regulated genes

      • Evaluation of cytotoxicity against cancer cell lines

      • Comparison with effects in HSF1-depleted cells to confirm specificity

    The development of compound I HSF115 illustrates this approach. Initial virtual screening identified compounds based on pharmacophoric criteria, followed by cell-based screening and SAR studies. I HSF115 was shown to bind the HSF1 DNA-binding domain and inhibit transcriptional activity without affecting oligomerization, nuclear localization, or DNA binding. The compound was used to probe the HSR at the transcriptome level and demonstrated cytotoxicity in various cancer cell lines, particularly multiple myeloma .

  • What methodologies can researchers use to study HSF1's mechanism of transcriptional regulation?

    Understanding HSF1's transcriptional regulatory mechanisms requires sophisticated methodological approaches:

    1. Chromatin immunoprecipitation (ChIP) approaches:

      • ChIP-seq to identify genome-wide HSF1 binding sites

      • ChIP-qPCR to quantify HSF1 occupancy at specific target genes

      • Sequential ChIP or Re-ChIP to identify co-occupancy with other factors

      • CUT&RUN or CUT&Tag as alternatives to traditional ChIP with potentially improved resolution

    2. Protein-protein interaction studies:

      • Co-immunoprecipitation to identify HSF1 interaction partners

      • Proximity ligation assays to detect protein interactions in situ

      • BioID or APEX proximity labeling to identify interaction networks

      • Yeast two-hybrid or mammalian two-hybrid assays for binary interactions

    3. Functional genomics approaches:

      • RNA-seq in HSF1-manipulated conditions to identify HSF1-dependent transcriptional programs

      • PRO-seq or GRO-seq to measure nascent transcription

      • ATAC-seq to assess chromatin accessibility changes mediated by HSF1

      • CRISPRi/a targeting HSF1 binding sites to assess functional relevance

    4. Single-cell approaches:

      • Single-cell RNA-seq to assess cell-to-cell variability in HSF1 responses

      • Single-cell ATAC-seq to examine chromatin accessibility changes

      • Live-cell imaging of fluorescently tagged HSF1 to observe dynamics

    Research has revealed that HSF1's transcriptional activity depends on multiple factors, including:

    • Recruitment to target promoters mediated by ATF1/CREB in response to stress

    • Interaction with the BRG1 chromatin-remodeling complex and p300/CBP

    • Participation in both gene activation and repression, as evidenced by I HSF115 studies showing that HSF1 mediates repression of heat-repressed genes

    • Interaction with other proteins including ATF1 and RPA1, which interact with the HSF1 DNA-binding domain

    These methodologies help distinguish between direct and indirect HSF1 effects and provide insights into the mechanisms by which HSF1 regulates transcription in different cellular contexts.

  • How do post-translational modifications (PTMs) impact HSF1 function and what experimental approaches can be used to study them?

    Post-translational modifications (PTMs) are critical regulators of HSF1 function. Research indicates that:

    • Phosphorylation of HSF1 at multiple sites affects its transcriptional activity

    • The phosphorylation status of HSF1 is one determinant of whether HSF1 trimers are transcriptionally competent

    Methodological approaches to study HSF1 PTMs include:

    1. Detection and mapping of PTMs:

      • Mass spectrometry-based proteomics to identify PTM sites and their dynamics

      • Phospho-specific antibodies to detect specific phosphorylation events

      • Phos-tag gels to separate phosphorylated from non-phosphorylated forms

      • 2D gel electrophoresis to visualize multiple HSF1 modification states

    2. Functional analysis of PTMs:

      • Site-directed mutagenesis to create phosphomimetic (e.g., S→D/E) or phospho-deficient (e.g., S→A) mutants

      • Expression of mutant forms in HSF1-depleted backgrounds to assess functional consequences

      • Pharmacological manipulation of kinases or phosphatases that modify HSF1

      • Temporal analysis of PTM dynamics during stress responses

    3. Identification of modifying enzymes:

      • Kinase inhibitor screens to identify kinases involved in HSF1 regulation

      • Co-immunoprecipitation followed by kinase assays to detect direct modification

      • Genetic screens to identify enzymes that affect HSF1 activity

    4. Integrated approaches:

      • Correlation of PTM status with HSF1 localization, DNA binding, and transcriptional activity

      • Systems biology approaches to model the impact of multiple PTMs on HSF1 function

      • Single-cell analysis to understand cell-to-cell variability in HSF1 modification states

    Understanding HSF1's PTMs is crucial as they may represent targetable nodes for therapeutic intervention, particularly in cancer contexts where HSF1 activity is dysregulated.

  • What is the HSF1 interactome and how do protein-protein interactions influence its function in different cellular contexts?

    HSF1 interacts with numerous proteins that influence its localization, stability, and transcriptional activity. The HSF1 interactome includes:

    • Chaperone proteins: HSP90 and co-chaperones that maintain HSF1 in an inactive state

    • Co-chaperones and regulators: CHIP, HDAC6, p97/VCP, DAXX, 14-3-3, FILIP-1L, and HSBP1

    • Transcriptional machinery components: ATF1/CREB, which mediate recruitment of HSF1 to target promoters

    • Chromatin remodelers: BRG1 complex and p300/CBP

    • DNA repair proteins: RPA1, which interacts with the HSF1 DNA-binding domain

    Methodological approaches to study the HSF1 interactome include:

    1. Identification of interaction partners:

      • Immunoprecipitation coupled with mass spectrometry (IP-MS)

      • Proximity-dependent biotinylation (BioID, APEX)

      • Yeast two-hybrid screens

      • Protein complementation assays (e.g., split luciferase)

    2. Validation and characterization of interactions:

      • Co-immunoprecipitation with specific antibodies

      • FRET or BRET to detect interactions in living cells

      • In vitro binding assays with recombinant proteins

      • Domain mapping to identify specific interaction regions

    3. Functional analysis of interactions:

      • Genetic or pharmacological disruption of specific interactions

      • Structure-function studies using deletion or point mutants

      • Temporal analysis of interaction dynamics during stress responses

      • Cell type-specific analysis to identify context-dependent interactions

    4. Therapeutic targeting of interactions:

      • Small molecule screens to identify compounds that disrupt specific interactions

      • Peptide-based approaches to compete with protein-protein interactions

      • Structure-based drug design targeting interaction interfaces

    The HSF1 inhibitor I HSF115 provides an example of how interactions can be targeted: while it does not affect heat-induced oligomerization, nuclear localization, or DNA binding, it inhibits the transcriptional activity of human HSF1 by interfering with the assembly of ATF1-containing transcription complexes . This demonstrates the potential of targeting specific protein-protein interactions within the HSF1 interactome for therapeutic purposes.

  • How do researchers evaluate HSF1 as a prognostic biomarker in cancer pathologies?

    Evaluating HSF1 as a prognostic biomarker requires rigorous methodological approaches to ensure reliability and clinical relevance:

    1. Patient cohort selection and sample processing:

      • Large, well-characterized patient cohorts with adequate follow-up (e.g., the Nurses' Health Study with 1,841 participants)

      • Standardized tissue processing and storage protocols

      • Construction of tissue microarrays for high-throughput analysis

    2. HSF1 detection and scoring:

      • Immunohistochemistry with validated anti-HSF1 antibodies

      • Scoring for nuclear HSF1 levels using standardized criteria

      • Automated image analysis to reduce subjective bias

      • Multi-observer scoring to ensure reproducibility

    3. Statistical analysis:

      • Kaplan-Meier analysis to assess associations with survival outcomes

      • Cox proportional hazard models to account for covariates

      • Multivariate analysis to determine independent prognostic value

      • Stratification by molecular subtypes (e.g., ER status in breast cancer)

    4. Validation approaches:

      • Independent validation cohorts to confirm findings

      • Analysis of public expression profiling data to correlate HSF1 mRNA levels with outcomes

      • Comparison with established prognostic markers

      • Assessment of HSF1 in combination with other biomarkers

    Research has demonstrated that:

    • Nuclear HSF1 levels are elevated in ~80% of in situ and invasive breast carcinomas

    • HSF1 expression is associated with high histologic grade, larger tumor size, and nodal involvement at diagnosis (P < 0.0001)

    • High HSF1 levels are independently associated with increased mortality (hazards ratio: 1.62; 95% CI: 1.21–2.17; P < 0.0013)

    • The prognostic impact is particularly strong in ER-positive breast cancers (hazards ratio: 2.10; 95% CI: 1.45–3.03; P < 0.0001)

    These findings suggest that HSF1 should be evaluated prospectively as an independent prognostic indicator, particularly in ER-positive breast cancer .

  • What experimental approaches can be used to distinguish between direct and indirect effects of HSF1 in gene regulation?

    Distinguishing between direct and indirect HSF1-mediated gene regulation is crucial for understanding its regulatory network. Researchers can employ several complementary approaches:

    1. Chromatin occupancy studies:

      • ChIP-seq to identify genome-wide HSF1 binding sites

      • ChIP-exo or ChIP-nexus for higher resolution binding site mapping

      • CUT&RUN or CUT&Tag as alternatives with potentially reduced background

      • Motif analysis to identify canonical and non-canonical HSF1 binding elements

    2. Nascent transcription analysis:

      • PRO-seq or GRO-seq to measure immediate transcriptional responses

      • 4sU-seq or BrU-seq to label and capture newly synthesized RNA

      • Time-course experiments to distinguish primary from secondary responses

    3. Genetic manipulation approaches:

      • Rapid HSF1 activation/inactivation systems (e.g., degron tags, chemical-genetic approaches)

      • CRISPR interference targeting specific HSF1 binding sites

      • HSF1 mutants that selectively affect specific functions

    4. Inhibitor-based approaches:

      • Use of HSF1 inhibitors like I HSF115 compared with transcription or translation inhibitors

      • Time-resolved analysis after inhibitor treatment

      • Comparison with effects in HSF1-depleted cells

    5. Integrative analysis:

      • Correlation of binding data with expression changes

      • Network analysis to identify direct targets and downstream effectors

      • Mathematical modeling to predict direct vs. indirect effects

    A particularly valuable approach is the use of chimeric transcription factors, as demonstrated in the development of I HSF115. Researchers created a stable cell line (Z74) reporting effects on both wild-type HSF1 and a chimeric HSF1 (with the HSF1 DNA-binding domain replaced by an unrelated DNA-binding domain). This allowed them to discriminate between compounds that directly target the HSF1 DNA-binding domain and those that affect HSF1 activity indirectly .

    Such approaches have revealed that while HSF1 directly regulates heat shock protein genes, it also influences a much broader transcriptional program, including both activation and repression of non-heat shock genes .

  • How can researchers study the tissue-specific and context-dependent functions of HSF1?

    HSF1 functions vary across tissues and cellular contexts, requiring specialized approaches to understand this diversity:

    1. Tissue-specific profiling:

      • Single-cell RNA-seq to identify cell type-specific HSF1-dependent gene expression

      • Cell type-specific ChIP-seq to map tissue-specific HSF1 binding patterns

      • Spatial transcriptomics to maintain tissue architecture context

      • Proteomics across tissues to identify tissue-specific HSF1 interactors

    2. Genetic approaches:

      • Tissue-specific HSF1 knockout or knockdown models

      • Conditional HSF1 expression systems in specific cell types

      • CRISPR-based screens in different cell types to identify context-dependent dependencies

      • Patient-derived models to capture disease-specific contexts

    3. Environmental and physiological contexts:

      • HSF1 activation under various stress conditions (heat, oxidative stress, etc.)

      • Cell cycle phase-specific analysis of HSF1 function

      • Metabolic state influence on HSF1 activity

      • Aging-related changes in HSF1 response

    4. Disease contexts:

      • Cancer vs. normal tissue comparisons

      • Neurodegenerative disease models

      • Inflammatory and stress conditions

      • Developmental contexts

    5. Multi-omics integration:

      • Correlation of transcriptome, proteome, and metabolome data

      • Epigenomic profiling across contexts

      • Network analysis to identify context-specific regulatory hubs

      • Systems biology approaches to model context-dependent behavior

    Research has shown that HSF1's role extends far beyond its classical heat shock response function, with implications in cancer progression, metabolism, gametogenesis, and aging . In cancer specifically, HSF1 has been shown to have context-dependent effects, with particularly strong prognostic significance in ER-positive breast cancers .

    Understanding these context-dependent functions may reveal new therapeutic opportunities by targeting HSF1 in specific disease contexts while potentially minimizing effects in normal tissues.

  • What computational and systems biology approaches can be used to study HSF1 regulatory networks?

    Systems biology and computational approaches offer powerful tools for understanding the complex regulatory networks involving HSF1:

    1. Network construction and analysis:

      • Inference of gene regulatory networks from transcriptomic data

      • Protein-protein interaction networks based on proteomics data

      • Integration of ChIP-seq and expression data to build directed networks

      • Network motif analysis to identify recurring regulatory patterns

    2. Dynamic modeling approaches:

      • Ordinary differential equation (ODE) models of HSF1 activation dynamics

      • Boolean network models of HSF1-dependent signaling

      • Stochastic models to capture cell-to-cell variability in HSF1 responses

      • Agent-based models to simulate multicellular HSF1-mediated responses

    3. Machine learning applications:

      • Prediction of HSF1 binding sites beyond canonical heat shock elements

      • Classification of direct vs. indirect HSF1 targets

      • Integration of multi-omics data to predict HSF1 activity

      • Identification of biomarkers for HSF1 activity in patient samples

    4. Comparative genomics approaches:

      • Cross-species analysis of HSF1-regulated genes

      • Evolutionary conservation of HSF1 binding sites

      • Identification of species-specific features of HSF1 regulation

    5. Drug discovery informatics:

      • Structure-based virtual screening for HSF1 inhibitors

      • Pharmacophore modeling based on known HSF1 binders

      • Prediction of compound effects on HSF1 networks

      • In silico modeling of combination therapies targeting HSF1-dependent pathways

    These approaches have facilitated discoveries such as:

    • Identification of four potential cavities in the HSF1 DNA-binding domain large enough to accommodate small drug-like molecules, leading to the development of HSF1 inhibitors

    • Recognition that HSF1 regulates a large majority of heat-induced genes and mediates repression of a significant fraction of heat-repressed genes

    • Understanding the extensive reprogramming of transcription by HSF1 beyond its classical heat shock protein targets

    Integration of experimental data with computational approaches continues to refine our understanding of HSF1's complex regulatory roles and identify new therapeutic opportunities.

  • How can researchers develop effective therapeutic strategies targeting HSF1 in cancer?

    Developing HSF1-targeted therapies requires a multifaceted approach:

    1. Target validation and patient stratification:

      • Identification of cancer types highly dependent on HSF1 activity

      • Biomarker development to identify HSF1-dependent tumors

      • Patient stratification strategies (e.g., focus on ER-positive breast cancers where HSF1 shows strong prognostic significance)

      • Synthetic lethality screening to identify context-dependent vulnerabilities

    2. Diverse inhibition strategies:

      • Direct inhibitors of the HSF1 DNA-binding domain (e.g., I HSF115)

      • Compounds targeting HSF1 activation (oligomerization, nuclear translocation)

      • Disruption of specific protein-protein interactions (e.g., HSF1-ATF1 complexes)

      • Degraders (PROTACs) targeting HSF1 for proteasomal degradation

    3. Combination therapy approaches:

      • HSF1 inhibitors with conventional chemotherapies

      • Combination with proteasome inhibitors to enhance proteotoxic stress

      • Targeting HSF1 along with key downstream effectors

      • Sequential therapy strategies to prevent resistance development

    4. Preclinical testing methodologies:

      • Cancer cell line panels to assess spectrum of activity

      • Patient-derived xenograft models to maintain tumor heterogeneity

      • Genetically engineered mouse models to assess efficacy and toxicity

      • Ex vivo organoid cultures for medium-throughput drug testing

    5. Monitoring therapeutic response:

      • Pharmacodynamic biomarkers of HSF1 inhibition (e.g., HSP70 expression)

      • Imaging approaches to assess tumor response

      • Liquid biopsy approaches to monitor treatment efficacy

      • Resistance mechanism identification through sequential sampling

    Research has shown that:

    • HSF1 inhibition through I HSF115 is cytotoxic for a variety of human cancer cell lines, with multiple myeloma lines consistently exhibiting high sensitivity

    • Genetic elimination of HSF1 protects mice from tumors induced by RAS oncogene mutations or P53 hot spot mutations

    • High nuclear HSF1 levels correlate with poor survival in breast cancer patients

    • HSF1 promotes the survival and proliferation of malignant cells

    These findings suggest that HSF1 may ultimately be a useful therapeutic target in multiple cancer types, with potential for both direct targeting and combination strategies to exploit cancer cells' dependence on HSF1-mediated transcriptional programs.

Product Science Overview

Introduction

Heat Shock Transcription Factor-1 (HSF1) is a highly conserved transcription factor found in eukaryotes. It plays a crucial role in the cellular response to stress, particularly heat shock, by regulating the expression of heat shock proteins (HSPs). These proteins function as molecular chaperones, aiding in the refolding of misfolded proteins and the degradation of damaged proteins. HSF1 is not only essential for stress response but also involved in various physiological processes, including development, metabolism, and aging .

Structure and Activation

HSF1 is characterized by an N-terminal helix-turn-helix DNA-binding domain and an adjacent oligomerization domain consisting of hydrophobic heptad repeats (HR-A/B). In unstressed cells, HSF1 exists in an inactive monomeric form. Upon exposure to stress, HSF1 undergoes trimerization and phosphorylation, which activates its DNA-binding ability. The activated HSF1 trimer translocates to the nucleus, where it binds to heat shock-responsive DNA elements (HSEs) to initiate the transcription of HSP genes .

Role in Stress Response

The primary function of HSF1 is to mediate the transcriptional response to proteotoxic stress. When cells are exposed to elevated temperatures or other stressors, HSF1 rapidly induces the expression of HSPs. These proteins help maintain protein homeostasis (proteostasis) by preventing the aggregation of misfolded proteins and facilitating their refolding or degradation. This response is critical for cell survival under stress conditions .

Involvement in Non-Stress Conditions

Recent studies have revealed that HSF1 also plays significant roles in non-stress conditions. It is involved in various physiological processes, including metabolism, gametogenesis, and aging. HSF1’s ability to reprogram transcription extends beyond the heat shock response, influencing a wide range of cellular functions. For instance, HSF1 has been implicated in cancer progression, where it supports the survival and proliferation of cancer cells by regulating the expression of genes involved in cell growth and survival .

Recombinant HSF1

Recombinant HSF1 refers to the HSF1 protein that has been produced using recombinant DNA technology. This involves inserting the HSF1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant HSF1 is used in various research applications to study its structure, function, and role in cellular processes. It is also employed in drug discovery efforts aimed at targeting HSF1 for therapeutic purposes, particularly in diseases where HSF1 activity is dysregulated .

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.