HSPBP1 Human

Heat Shock Protein-Binding Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of HSPBP1 Human

HSPBP1 (Heat shock protein 70-binding protein 1) is a eukaryotic protein that regulates HSP70 activity through nucleotide exchange and inhibition of ATP hydrolysis. It is expressed predominantly in heart, skeletal muscle, and testis tissues . HSPBP1 interacts with HSP70’s ATPase domain, preventing ATP binding and modulating its chaperone functions . Its dual role as a tumor suppressor and regulator of protein quality control makes it critical in both cancer biology and neurodegenerative diseases.

Molecular Structure and Properties

PropertyDetailSource
Molecular Mass41.6 kDa (recombinant form with His-Tag)
Expression TissuesHeart, skeletal muscle, testis; lower in astrocytes compared to neurons
LocalizationCytoplasmic, nuclear, and extracellular
FormulationTris-HCl buffer (pH 8.0), 0.1M NaCl, 30% glycerol, 2mM DTT, 2mM EDTA

HSPBP1 is a non-glycosylated polypeptide (382 amino acids) produced via recombinant expression in E. coli . Its N-terminal His-Tag facilitates purification .

HSP70 Regulation

HSPBP1 binds to HSP70 (e.g., HSPA1A, HSPA8) and inhibits ATPase activity by 90%, disrupting nucleotide exchange . This modulation impacts:

  • Chaperone Activity: Reduces HSP70’s ability to refold misfolded proteins .

  • Apoptosis Suppression: Inhibits HSP70-mediated anti-apoptotic signaling, promoting cell death under stress .

CHIP Inhibition

HSPBP1 antagonizes STUB1 (CHIP), an E3 ubiquitin ligase that targets misfolded proteins for degradation. In neurons, high HSPBP1 levels suppress CHIP activity, impairing protein quality control and exacerbating neurodegenerative pathologies .

Partner ProteinInteraction MechanismFunctional Outcome
HSPA1ABinds ATPase domain, blocks ATP bindingInhibits chaperone activity
STUB1Competes with CHIP for HSP70 bindingReduces ubiquitination of misfolded proteins
BRCA1Promotes BRCA1-mediated DNA repairEnhances genomic stability in breast cancer

Cancer: Tumor Suppression and Progression

  • Breast Cancer:

    • Tumor Suppression: HSPBP1 interacts with BRCA1 to enhance homologous recombination repair, reducing genomic instability .

    • Apoptosis Promotion: Inhibits HSP70’s survival signaling, sensitizing cells to stress .

  • Hepatocellular Carcinoma (HCC):

    • Upregulation: Elevated HSPBP1 levels correlate with HSP70 overexpression in HCV-related HCC .

    • Prognostic Marker: Lower HSPBP1:HSP70 ratio in larger tumors suggests aggressive progression .

Neurodegeneration: Neuron vs. Astrocyte Vulnerability

HSPBP1 expression is 5-fold higher in neurons than astrocytes, leading to:

  • CHIP Inhibition: Reduced protein degradation in neurons, causing misfolded protein accumulation (e.g., huntingtin in Huntington’s disease) .

  • Therapeutic Targeting: Knocking down HSPBP1 in neurons restores CHIP activity, clears aggregates, and reduces neuropathology .

Reproductive Health: Male Fertility

HSPBP1 deficiency in mice causes male sterility due to apoptosis of spermatocytes. It stabilizes inducible HSP70 isoforms, protecting germ cells from proteotoxic stress .

Product Specs

Introduction
Hsp70-binding protein 1 (HSPBP1) belongs to a family of eukaryotic proteins known as nucleotide exchange factors for HSP 70. These proteins demonstrate varying degrees of specificity for compartments and species. HSPBP1 is primarily found in the cytoplasm and nucleus, but it can also be detected outside of cells. Its expression is highest in heart and skeletal muscle. HSPBP1 interacts with HSP 70, inhibiting its activity and facilitating the release of nucleotides from the ATPase domain of HSP 70. Moreover, HSPBP1 hinders the chaperone activity of HSPA1A by altering the conformation of its ATP-binding domain, thereby impeding ATP binding. There is a possibility that HSPBP1 plays a role in the (dys)regulation of chaperone proteins in tumors. Additionally, HSPBP1 interferes with STUB1-mediated ubiquitination and hampers the chaperone-assisted degradation of immature CFTR.
Description
Recombinant human HSPBP1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 382 amino acids (specifically, amino acids 1 to 362). It possesses a molecular weight of 41.6 kDa. A 20 amino acid His Tag is fused to the N-terminus of HSPBP1. The protein undergoes purification using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
HSPBP1 solution is prepared in a buffer containing 20mM Tris-HCl (pH 8.0), 2mM DTT, 30% glycerol, 2mM EDTA, and 0.1M NaCl.
Stability
Recombinant human HSPBP1 maintains stability for 1 week when stored at 4°C. However, it is recommended to store the protein at a temperature below -18°C to ensure optimal long-term stability. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the protein is determined to be greater than 95% based on SDS-PAGE analysis.
Synonyms
Hsp70-binding protein 1, Heat shock protein-binding protein 1, Hsp70-interacting protein 1, Hsp70-binding protein 2, Hsp70-interacting protein 2, HspBP1, HspBP2, HSPBP1, HSPBP, FES1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSDEGSRGSR LPLALPPASQ GCSSGGGGGG GGGSSAGGSG NSRPPRNLQG LLQMAITAGS EEPDPPPEPM SEERRQWLQE AMSAAFRGQR EEVEQMKSCL RVLSQPMPPT AGEAEQAADQ QEREGALELL ADLCENMDNA ADFCQLSGMH LLVGRYLEAG AAGLRWRAAQ LIGTCSQNVA AIQEQVLGLG ALRKLLRLLD RDACDTVRVK ALFAISCLVR EQEAGLLQFL RLDGFSVLMR AMQQQVQKLK VKSAFLLQNL LVGHPEHKGT LCSMGMVQQL VALVRTEHSP FHEHVLGALC SLVTDFPQGV RECREPELGL EELLRHRCQL LQQHEEYQEE LEFCEKLLQT CFSSPADDSM DR.

Q&A

What is HSPBP1 and what is its primary function in cellular systems?

HSPBP1 (also known as Hsp70-binding protein 1) is a cochaperone protein that primarily regulates the activity of HSP70 family proteins. It functions by inhibiting HSPA1A chaperone activity through altering the conformation of the ATP-binding domain of HSPA1A, which interferes with ATP binding . HSPBP1 also plays a critical role in protein quality control by interfering with ubiquitination mediated by STUB1 (also known as CHIP) and inhibits chaperone-assisted degradation of proteins like immature CFTR . This regulatory function positions HSPBP1 as a key modulator in the protein homeostasis network, where it helps determine the fate of client proteins bound to HSP70 chaperones. The protein exerts prosurvival functions by inhibiting the ubiquitylation and proteasomal degradation of inducible HSP70 family members . Methodologically, these functions have been elucidated using recombinant protein studies, co-immunoprecipitation assays, and knockout mouse models.

What are the expression patterns of HSPBP1 across human tissues?

HSPBP1 displays differential expression across tissues, with particularly high expression in testis. Immunoblot analyses of tissue extracts reveal weak to moderate expression in brain, muscle, colon, and small intestine but substantially stronger expression in testicular tissue . This tissue-specific expression pattern suggests specialized roles in reproductive tissues. In situ hybridization experiments have demonstrated that HSPBP1 mRNA is specifically expressed in cells within seminiferous tubules of adult testes . When examining neural tissues specifically, HSPBP1 shows markedly higher expression in neurons compared to astrocytes, which has significant implications for neurodegeneration and protein misfolding diseases . Researchers investigating HSPBP1 should consider these tissue-specific variations when designing experiments and interpreting results.

What techniques are most effective for detecting and measuring HSPBP1 in experimental settings?

For effective detection and quantification of HSPBP1 in research settings, several complementary techniques should be considered:

  • Immunoblotting (Western blot): Using specific anti-HSPBP1 antibodies has proven reliable for protein detection and semi-quantitative analysis across tissue lysates, as demonstrated in studies comparing expression across multiple organs .

  • In situ hybridization: This technique effectively localizes HSPBP1 mRNA expression at the cellular level, particularly valuable for heterogeneous tissues such as testes, where expression can be mapped to specific cell types within the seminiferous tubules .

  • Immunohistochemistry/Immunofluorescence: For spatial localization of the protein within tissues and cells.

  • qRT-PCR: For quantitative assessment of HSPBP1 transcript levels across different experimental conditions.

  • RNA-seq: Used for comparative transcriptomic profiling, as evidenced by studies identifying differential HSPBP1 expression between neurons and astrocytes .

Each method provides distinct advantages, with selection depending on specific research questions. For cellular localization studies, combined approaches using both protein and mRNA detection provide more comprehensive insights into HSPBP1 biology.

How does HSPBP1 regulate the protein quality control system through its interaction with the CHIP/Hsp70 complex?

HSPBP1 exerts sophisticated control over protein quality control through multiple molecular mechanisms. The protein directly inhibits the E3 ubiquitin ligase activity of CHIP (C terminus of Hsp70-interacting protein), a key cochaperone that determines the fate of HSP70-bound client proteins . This inhibition occurs through competitive binding to the HSP70 chaperone, preventing CHIP from facilitating K48-linked polyubiquitination that would otherwise target misfolded proteins for proteasomal degradation .

Research has demonstrated that HSPBP1 affects chaperone expression at a posttranslational level by specifically inhibiting the ubiquitylation and proteasomal degradation of inducible HSP70 proteins . This creates a feedback loop whereby HSPBP1 not only regulates client protein degradation but also stabilizes the chaperones themselves. The complex interplay between these factors has been elucidated through biochemical assays measuring ubiquitination rates, protein stability assays using cycloheximide chase experiments, and co-immunoprecipitation studies demonstrating differential binding affinities.

Notably, the activity of CHIP shows marked differences between cell types, being more active in astrocytes than neurons due to differential HSPBP1 expression . In astrocytes with lower HSPBP1 levels, CHIP is more actively monoubiquitinated and binds to misfolded proteins (such as mutant huntingtin) more avidly, facilitating their clearance . This mechanistic understanding has emerged from sophisticated proteomics approaches and cellular models of protein misfolding diseases.

What is the role of HSPBP1 in neurodegeneration and why does it contribute to neuronal vulnerability?

HSPBP1 plays a critical role in determining the differential vulnerability of neurons versus glial cells to neurodegenerative processes. Neurons express significantly higher levels of HSPBP1 compared to astrocytes (approximately 5-fold higher according to RNA-seq data), which directly impacts their ability to clear misfolded proteins . This abundant neuronal HSPBP1 expression inhibits CHIP E3 ligase activity, resulting in reduced degradation of misfolded proteins that are characteristic of neurodegenerative diseases.

The mechanistic consequence of elevated HSPBP1 in neurons includes:

Experimental evidence supporting this model comes from studies showing that silencing HspBP1 expression via CRISPR-Cas9 in neurons ameliorated mHtt aggregation and neuropathology in HD knockin mouse models . This suggests that HSPBP1 inhibition could represent a novel therapeutic target for neurodegenerative diseases.

The research methodology advancing this understanding has involved comparative cellular models, transgenic mouse studies, and advanced proteomics to characterize the differential CHIP/Hsp70 activities in neuronal and glial cells. These findings provide a molecular explanation for the long-standing observation that neurons are more vulnerable than glia to misfolded protein accumulation in various neurodegenerative diseases.

What is the significance of HSPBP1 in spermatogenesis and male fertility?

HSPBP1 plays an essential role in spermatogenesis and male fertility, as evidenced by targeted gene disruption studies. Male HSPBP1<sup>−/−</sup> mice are sterile due to impaired meiosis and massive apoptosis of spermatocytes . The molecular basis for this phenotype involves HSPBP1's regulation of specific HSP70 family members crucial for spermatogenesis.

HSPBP1 deficiency in testes strongly reduces the expression of inducible, antiapoptotic HSP70 family members, particularly HSPA1L and HSPA2 . HSPA2 is essential for synaptonemal complex disassembly during meiosis, explaining why its loss leads to meiotic arrest. Histological examination of HSPBP1<sup>−/−</sup> testes reveals a severe reduction in germ cells within seminiferous tubules, while epididymides are completely devoid of mature spermatozoa .

The mechanism behind this phenotype involves post-translational regulation. HSPBP1 stabilizes inducible HSP70 proteins by preventing their ubiquitination and proteasomal degradation . Without this protection, these crucial chaperones are degraded, compromising spermatocyte survival.

Methodologically, this has been investigated through:

  • Targeted gene disruption techniques to generate HSPBP1 knockout mice

  • TUNEL assays demonstrating increased apoptosis in seminiferous tubules

  • Immunohistochemical analysis showing reduction in specific HSP70 family members

  • Detailed staging of meiotic progression using markers like SYCP3 and histone H1t

  • Assessment of chromosome synapsis and recombination through MLH1 foci quantification

These findings establish HSPBP1 as a critical posttranslational regulator of chaperone abundance in the testis and highlight its essential role in male fertility.

What genetic models are available for studying HSPBP1 function and what are their key phenotypes?

Several genetic models have been developed to study HSPBP1 function, each offering unique insights into its biological roles:

Transgenic Mouse Models:

  • HSPBP1 Knockout (HSPBP1<sup>−/−</sup>): Generated through targeted disruption of the mouse HSPBP1 locus. Key phenotypes include:

    • Male sterility due to impaired spermatogenesis

    • Normal female fertility

    • Reduced testis weight (approximately 50% reduction by 8 weeks)

    • Absence of mature spermatozoa in epididymides

    • Increased apoptosis in seminiferous tubules

    • Normal body weight and development of other organs

  • Conditional Neuronal HSPBP1 Knockdown: Using CRISPR-Cas9 technology for targeted silencing in neurons. Phenotypes include:

    • Reduced aggregation of mutant huntingtin protein

    • Ameliorated neuropathology in HD knockin mouse brains

    • Enhanced CHIP-mediated degradation of misfolded proteins

Cellular Models:

  • Primary neurons vs. astrocyte cultures: Valuable for studying differential HSPBP1 expression and its consequences for protein quality control

  • HSPBP1-overexpression systems: Used to assess gain-of-function effects on chaperone networks and proteostasis

  • HSPBP1 siRNA/shRNA knockdown cell lines: Employed for acute depletion studies

These models have revealed that while HSPBP1 is essential for male fertility through its role in spermatogenesis, it may have detrimental effects in neurons by inhibiting clearance of misfolded proteins. The availability of these complementary models allows researchers to study tissue-specific functions and explore therapeutic potential in neurodegenerative conditions.

What experimental approaches can be used to modulate HSPBP1 activity in disease models?

Several experimental approaches have been developed to modulate HSPBP1 activity in disease models, particularly focusing on neurodegenerative conditions:

Genetic Approaches:

  • CRISPR-Cas9 gene editing: Successfully employed to silence HspBP1 expression in neurons, resulting in ameliorated mutant huntingtin aggregation and improved neuropathology in HD knockin mouse models . This approach provides specific targeting with minimal off-target effects.

  • RNA interference: siRNA and shRNA constructs targeting HSPBP1 offer flexible options for temporal control of knockdown, suitable for both in vitro and in vivo applications.

  • Viral vector-mediated delivery: AAV or lentiviral systems can be used to deliver HSPBP1-targeting constructs or overexpression cassettes to specific brain regions or cell types.

Pharmacological Approaches:

  • Small molecule inhibitors: Although specific HSPBP1 inhibitors are still under development, high-throughput screening approaches are being utilized to identify compounds that disrupt HSPBP1-HSP70 interactions.

  • Indirect modulation: Targeting upstream regulators of HSPBP1 expression or post-translational modifications that affect its activity.

Methodological Considerations:
When implementing these approaches, researchers should monitor:

  • Changes in CHIP ubiquitination activity

  • Alterations in HSP70 levels and stability

  • Effects on clearance of disease-associated misfolded proteins

  • Cell-type specific responses, particularly between neurons and glia

  • Potential compensatory mechanisms involving other cochaperones like BAG2

The choice of approach depends on the specific research question, disease model, and desired temporal control of HSPBP1 modulation. Combined approaches may offer synergistic benefits and more comprehensive insights into HSPBP1's therapeutic potential in neurodegenerative diseases.

How can researchers effectively measure HSPBP1-mediated effects on protein degradation pathways?

Measuring HSPBP1's effects on protein degradation requires multiple complementary approaches that address both client protein fate and chaperone dynamics:

Ubiquitination Assays:

  • In vitro ubiquitination: Using purified components (CHIP, E1, E2, ubiquitin, ATP, and substrate) with or without recombinant HSPBP1 to directly measure its inhibitory effect on CHIP-mediated ubiquitination.

  • Cellular ubiquitination: Immunoprecipitation of target proteins (e.g., misfolded clients or HSP70 itself) followed by ubiquitin immunoblotting to detect K48-linked polyubiquitination, which is typically reduced in the presence of HSPBP1 .

Protein Degradation Measurements:

  • Cycloheximide chase assays: To measure protein half-life differences in the presence or absence of HSPBP1.

  • Pulse-chase experiments: Using metabolic labeling to track the fate of newly synthesized proteins under different HSPBP1 conditions.

  • Fluorescent reporters: Utilizing destabilized GFP fused to model substrates to monitor degradation kinetics in real-time.

Chaperone Activity Assessments:

  • ATP binding/hydrolysis assays: To measure how HSPBP1 affects the ATPase activity of HSP70, which is crucial for client processing.

  • Client refolding assays: Using model substrates like luciferase to assess how HSPBP1 modulates HSP70-mediated protein refolding.

Aggregate Clearance Measurements:

  • Filter trap assays: To quantify changes in protein aggregation when HSPBP1 levels are modulated.

  • Fluorescence microscopy: To visualize and quantify aggregates of fluorescently tagged disease proteins (e.g., mutant huntingtin) under different HSPBP1 conditions .

When implementing these approaches, researchers should consider cell-type specific differences, as HSPBP1 effects are notably different between neurons and astrocytes . Additionally, compensatory mechanisms involving other cochaperones like BAG2 should be monitored, as they may contribute to HSP70 stabilization in certain tissues .

What are the emerging technologies for studying HSPBP1 interactions and functions?

Emerging technologies are revolutionizing our understanding of HSPBP1 biology by providing unprecedented resolution of its interactions and functions:

Proximity-Based Interaction Mapping:

  • BioID/TurboID: These proximity labeling techniques use HSPBP1 fused to a biotin ligase to identify proteins in its native microenvironment, revealing previously unknown interaction partners beyond the well-established HSP70-CHIP axis.

  • APEX2 proximity labeling: Offers temporal resolution for capturing dynamic HSPBP1 interactions under different cellular stresses.

Advanced Imaging Approaches:

  • Super-resolution microscopy: Techniques like STORM and PALM enable visualization of HSPBP1 subcellular localization and co-localization with partners at nanometer resolution.

  • Live-cell FRET/BRET sensors: Allow real-time monitoring of HSPBP1 interactions with HSP70 and other partners in living cells under various conditions.

Structural Biology Advances:

  • Cryo-EM: Provides detailed structural information on HSPBP1-HSP70-client complexes, informing mechanism-based therapeutic design.

  • HDX-MS (Hydrogen-Deuterium Exchange Mass Spectrometry): Maps conformational changes in HSP70 induced by HSPBP1 binding, revealing allosteric mechanisms.

Single-Cell Technologies:

  • Single-cell transcriptomics: Reveals cell-type specific expression patterns of HSPBP1 and its partners in complex tissues like brain and testis.

  • Single-cell proteomics: Emerging techniques allow measurement of HSPBP1 protein levels and modification states at single-cell resolution.

Genome Engineering:

  • CRISPR base editing and prime editing: Enables precise manipulation of HSPBP1 at the genomic level to model disease variants or create functional mutations.

  • CRISPR interference/activation (CRISPRi/CRISPRa): Allows temporal and reversible modulation of HSPBP1 expression without permanent genetic changes.

These technologies are particularly valuable for understanding the cell-type specific roles of HSPBP1 in the differential vulnerability of neurons versus astrocytes to protein misfolding diseases and its critical function in spermatogenesis .

How might targeting HSPBP1 serve as a therapeutic strategy for neurodegenerative diseases?

Targeting HSPBP1 represents a promising therapeutic strategy for neurodegenerative diseases based on its role in regulating protein quality control. The rationale stems from several key observations:

  • Differential expression pattern: Neurons express significantly higher levels of HSPBP1 than astrocytes, which correlates with their greater vulnerability to protein aggregation diseases .

  • Inhibitory effect on protein clearance: HSPBP1 inhibits CHIP E3 ligase activity, thereby reducing the degradation of misfolded proteins that characterize neurodegenerative diseases .

  • Proof-of-concept studies: Silencing HspBP1 expression via CRISPR-Cas9 in neurons successfully ameliorated mutant huntingtin (mHtt) aggregation and neuropathology in HD knockin mouse brains .

Potential therapeutic approaches include:

  • Antisense oligonucleotides (ASOs): These could be designed to reduce HSPBP1 expression with high specificity. ASO therapy has already shown promise in other neurodegenerative conditions.

  • Small molecule inhibitors: Compounds that disrupt the interaction between HSPBP1 and HSP70 could restore CHIP activity and enhance clearance of misfolded proteins.

  • Peptide-based inhibitors: Designed to mimic critical binding interfaces between HSPBP1 and its partners.

  • Viral vector-mediated gene therapy: Using AAV or other vectors to deliver HSPBP1-targeting constructs specifically to neurons in affected brain regions.

Methodological considerations for therapeutic development:

  • Target validation: Further studies in diverse neurodegenerative disease models (Alzheimer's, Parkinson's) are needed to validate HSPBP1 as a broad-spectrum target.

  • Cell-type specificity: Strategies should ideally target neurons while sparing HSPBP1 function in other tissues where it plays beneficial roles (e.g., testes) .

  • Timing of intervention: Determining optimal therapeutic windows for HSPBP1 targeting in disease progression.

  • Biomarkers: Developing measures of target engagement and efficacy, possibly by monitoring levels of misfolded proteins or CHIP activity.

The therapeutic potential of HSPBP1 targeting is particularly promising because it addresses a fundamental aspect of neurodegeneration—impaired protein quality control—rather than targeting disease-specific proteins, potentially offering broad applicability across multiple neurodegenerative conditions.

What are the key considerations for developing experimental protocols to study HSPBP1 in human samples?

Developing robust experimental protocols for studying HSPBP1 in human samples requires careful consideration of several critical factors:

Sample Collection and Processing:

  • Tissue specificity: Given HSPBP1's differential expression across tissues (high in testis and neurons, lower in astrocytes and other tissues) , selection of appropriate sample types is crucial. For neurodegenerative studies, both affected and unaffected brain regions should be compared.

  • Post-mortem considerations: For brain tissue, post-mortem interval significantly affects protein quality and modifications. Standardized protocols with rapid fixation/freezing are essential.

  • Preservation methods: Optimization for different analytical techniques (e.g., protein vs. RNA analysis, immunohistochemistry vs. biochemical assays).

Analytical Approaches:

  • Protein detection optimization: HSPBP1 detection requires validated antibodies specifically tested in human tissues. Western blotting protocols should be optimized for human sample heterogeneity.

  • Cell-type specific analyses: Given the significant differences between neurons and glia , techniques like laser capture microdissection or single-cell approaches are valuable for isolating specific cell populations from human brain tissues.

  • Activity assays: Beyond measuring HSPBP1 levels, functional assays to assess its inhibitory activity on CHIP-mediated ubiquitination in human samples provide more meaningful insights.

Clinical Correlation:

  • Patient stratification: Careful documentation of clinical parameters, disease stage, and concurrent pathologies for correlation with HSPBP1 findings.

  • Genetic analysis: Screening for HSPBP1 variants or expression quantitative trait loci (eQTLs) that might influence disease susceptibility.

  • Biobanking considerations: Standardized protocols for sample collection, processing, and storage to enable reliable multi-center studies.

Ethical Considerations:

  • Informed consent: Especially important for fertility-related studies given HSPBP1's role in spermatogenesis .

  • Incidental findings: Protocols for handling potentially clinically relevant discoveries, particularly in genetic studies.

These methodological considerations are essential for translating the fundamental discoveries about HSPBP1 from animal and cell models to human disease contexts, ultimately advancing its potential as a therapeutic target for neurodegenerative conditions.

Product Science Overview

Introduction

Heat Shock Protein-Binding Protein 1 (HSBP1) is a crucial component in the cellular response to stress. It is a 76-amino-acid protein that binds to Heat Shock Factor 1 (HSF1), a transcription factor involved in the heat shock response. This response is triggered by exposure to thermal and chemical stress, leading to the elevated expression of heat-shock-induced genes .

Structure and Function

HSBP1 is nuclear-localized and interacts with the active trimeric state of HSF1 to negatively regulate HSF1 DNA-binding activity . During the heat shock response, HSF1 undergoes a conformational transition from an inert non-DNA-binding monomer to active functional trimers . HSBP1 binds to these trimers, repressing the transactivation activity of HSF1 .

Role in Cellular Stress Response

The heat shock response is a protective mechanism that helps cells survive under adverse conditions such as elevated temperatures, toxins, and other stressors. HSBP1 plays a negative regulatory role in this process. Overexpression of HSBP1 in mammalian cells represses the transactivation activity of HSF1, thereby modulating the heat shock response . In model organisms like C. elegans, overexpression of HSBP1 has severe effects on survival after thermal and chemical stress, consistent with its role as a negative regulator .

Implications in Aging and Disease

HSBP1 may also have a role in the suppression of the activation of the stress response during the aging process . Its interaction with HSF1 and the subsequent regulation of heat shock proteins (HSPs) are crucial for maintaining cellular homeostasis. Dysregulation of HSBP1 and HSF1 activity has been implicated in various diseases, including cancer .

Recombinant HSBP1

Recombinant HSBP1 is produced using recombinant DNA technology, which involves inserting the HSBP1 gene into an expression vector and introducing it into a host cell, such as E. coli or yeast. The host cells then produce the HSBP1 protein, which can be purified for research and therapeutic purposes. Recombinant HSBP1 is used in various studies to understand its function and role in the heat shock response and other cellular processes.

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.