HSPB2 Human

Heat Shock 27kDa Protein 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

HSPB2 is encoded by the HSPB2 gene located on chromosome 11 (11q23.3) in humans . Key structural and molecular features include:

Biological Functions

HSPB2 exhibits diverse roles in cellular maintenance and stress adaptation:

Table 2: Functional Roles of HSPB2

FunctionMechanismReferences
Chaperone ActivityPrevents aggregation of misfolded proteins (e.g., insulin, α-synuclein) under stress
DMPK RegulationActivates Myotonic Dystrophy Protein Kinase (DMPK) to maintain muscle integrity
Autophagy ModulationPromotes clearance of damaged proteins via BAG3/SQSTM1 complex in neurons
OligomerizationForms dynamic, concentration-dependent oligomers; exchanges subunits with HSPB3

Disease Associations

HSPB2 dysregulation is linked to multiple pathologies:

Table 3: Clinical Implications of HSPB2

DiseaseRole of HSPB2References
Myotonic DystrophyBinds DMPK; upregulated in patient muscle tissues
Traumatic Brain Injury (TBI)Enhances neural regeneration via autophagy; improves long-term sensorimotor recovery
CancerDownregulated in breast carcinoma; overexpression correlates with poor prognosis
Cardiac IschemiaProtects against ATP depletion during ischemia/reperfusion injury

Research Findings

Key studies highlight HSPB2’s mechanistic and therapeutic relevance:

  1. Structural Insights:

    • HSPB2 adopts a β-sheet-rich conformation with solvent-exposed hydrophobic surfaces .

    • Oligomerizes dynamically, facilitating interactions with HSPB3 and other sHSPs .

  2. Neuroprotection:

    • In TBI models, HSPB2 overexpression accelerates white matter repair and neuronal plasticity .

    • Autophagy inhibition (e.g., chloroquine) blocks its neuroregenerative effects .

  3. Cancer Dynamics:

    • HSPB2 silencing enhances chemosensitivity in bladder cancer cells, increasing apoptosis .

    • Low HSPB2 mRNA levels in breast cancer correlate with tumor progression .

Expression Patterns

HSPB2 is predominantly expressed in cardiac and skeletal muscle, with stress-inducible upregulation in neurons . Key expression changes include:

Table 4: HSPB2 Expression Under Pathological Conditions

ConditionExpression ChangeReferences
Myotonic DystrophyUpregulated in muscle tissues
TBITransient increase in cortical and hippocampal neurons post-injury
Breast CancerDownregulated in tumor tissues vs. non-cancerous counterparts

Therapeutic Potential

Emerging strategies targeting HSPB2 include:

  • Autophagy Enhancement: Acute-stage HSPB2 induction improves TBI outcomes .

  • Chemosensitization: HSPB2/HSPB3 knockdown synergizes with doxorubicin in bladder cancer .

  • Metabolic Regulation: Cardiac-specific HSPB2 overexpression mitigates ischemic damage .

Product Specs

Introduction
Heat shock protein beta-2 (HSPB2), a member of the small heat shock protein (HSP20) family, regulates DMPK kinase.
Description
Recombinant HSPB2 Human, produced in E. coli, is a single polypeptide chain of 206 amino acids (1-182) with a molecular weight of 22.8kDa. It consists of amino acids 1-182 of HSPB2 fused to a 24 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
HSPB2 is supplied as a 0.5mg/ml solution in 20mM Tris-HCl buffer (pH 8.0), 1mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), store 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. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Heat shock 27kDa protein 2, DMPK-binding protein, MKBP, HSP27, LOH11CR1K.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMSGRSV PHAHPATAEY EFANPSRLGE QRFGEGLLPE EILTPTLYHG YYVRPRAAPA GEGSRAGASE LRLSEGKFQA FLDVSHFTPD EVTVRTVDNL LEVSARHPQR LDRHGFVSRE FCRTYVLPAD VDPWRVRAAL SHDGILNLEA PRGGRHLDTE VNEVYISLLP APPDPEEEEE AAIVEP.

Q&A

What is the structural organization of HSPB2 and how does it compare to other small heat shock proteins?

HSPB2 belongs to the family of small heat shock proteins (sHsps) consisting of ten members (HSPB1-HSPB10). Like other family members, HSPB2 possesses a highly conserved centrally located α-crystallin domain (ACD) and less conserved N- and C-terminal domains . Unlike some other sHsps, HSPB2 can form heteromeric complexes, most notably with HSPB3 in a 3:1 ratio. Structurally, the HSPB2/B3 heterotetramer shows a distinctive arrangement where the four α-crystallin domains assemble into a flattened tetrahedron pierced by two non-intersecting approximate dyads . This assembly is mediated by flexible "nuts and bolts" involving IXI/V motifs from terminal regions filling ACD pockets. Parts of the N-terminal region bind in an unfolded conformation into the anti-parallel shared ACD dimer grooves, while certain tracts of the terminal regions remain disordered, contributing to the protein's functional plasticity .

What is the tissue distribution pattern of HSPB2 and how is it regulated?

Unlike some ubiquitously expressed heat shock proteins, HSPB2 demonstrates a distinct tissue-specific expression pattern. It is preferentially expressed in the heart and skeletal muscle . During development, HSPB2 is transiently upregulated in the neonatal myocardium, suggesting a role in developmental processes . Interestingly, HSPB2 gene is closely linked to the αB-crystallin gene, which may imply coordinated regulation .

How does HSPB2 function as a chaperone and what are its target specificity patterns?

HSPB2 exhibits chaperone-like activity that appears to be target protein-dependent . Like other small heat shock proteins, HSPB2 acts as a holdase, interacting with substrate proteins to prevent their aggregation during stress conditions . A significant finding is that HSPB2 can effectively prevent the ordered amyloid fibril formation of α-synuclein, suggesting a potential role in preventing protein aggregation disorders like Parkinson's disease .

Research indicates that HSPB2 cooperates in substrate refolding driven by other chaperones or, alternatively, can promote substrate routing to degradation pathways . This functionality is crucial for maintaining proteostasis and structural integrity of the cytoskeleton. Additionally, HSPB2 plays an important role in ATP turnover in mouse heart and is associated with mitochondria, suggesting involvement in cellular energy metabolism .

What are the molecular mechanisms by which HSPB2 regulates DMPK activity and how does this relate to myotonic dystrophy?

HSPB2, also known as Myotonic Dystrophy Protein Kinase Binding Protein (MKBP), specifically associates with and activates Myotonic Dystrophy Protein Kinase (DMPK) . DMPK is a serine/threonine protein kinase essential for maintaining muscle structure and function. The interaction between HSPB2 and DMPK likely involves direct binding that influences DMPK's kinase activity.

Myotonic dystrophy (DM) is associated with mutations in the DMPK gene, leading to RNA toxicity. The HSPB2-DMPK interaction suggests that HSPB2 may serve as a regulator of DMPK function, potentially influencing disease progression . The enzyme activator activity noted in Gene Ontology annotations for HSPB2 likely refers to this DMPK activation function .

Research approaches to study this interaction should include co-immunoprecipitation assays, kinase activity assays in the presence and absence of HSPB2, and structural studies of the HSPB2-DMPK complex. Additionally, investigating how disease-associated mutations in either protein affect their interaction could provide valuable insights into pathological mechanisms.

What role does HSPB2 play in neuroprotection and neural regeneration following traumatic brain injury?

Recent research has revealed a significant role for HSPB2 in neuroprotection and neural regeneration following traumatic brain injury (TBI). HSPB2 is markedly increased in neurons after TBI in both mouse models and human patients . This upregulation appears to be a universal phenomenon in acute neuronal injury, as it also occurs in in vitro models of oxygen-glucose deprivation followed by reperfusion (OGD/r) .

Using a tamoxifen-induced neuron-specific HSPB2 overexpression transgenic mouse model, researchers demonstrated that elevated HSPB2 levels promote long-term sensorimotor recovery and alleviate tissue loss after TBI . Specifically, HSPB2 enhances white matter structural and functional integrity, promotes central nervous system (CNS) plasticity, and accelerates long-term neural remodeling.

Functional studies showed that HSPB2 overexpression improves sensorimotor outcomes in multiple behavioral tests, including the body curl test, rotarod test, and grid-walking test . Additionally, HSPB2 confers partial protection against the loss of white matter integrity by improving the electrical conduction of unmyelinated axonal fibers and partially reverses the reduction in synaptic density (measured by synaptophysin) in the peri-injury cortex .

How does HSPB2 contribute to cardiac protection during ischemic stress?

HSPB2 plays a significant role in cardiac protection during ischemic stress through several mechanisms. During ischemic stress, HSPB2 translocates from the cytosolic fraction to the myofibril fraction of rat heart . This translocation is likely a protective response mechanism. Studies have shown that HSPB2 protects the myocardium from ischemia and helps in recovery during reperfusion .

The cardioprotective function of HSPB2 may be related to its importance in ATP turnover in mouse heart . By maintaining energy homeostasis during ischemic conditions, HSPB2 could prevent the deleterious effects of ATP depletion. Additionally, HSPB2's association with mitochondria suggests it may play a role in preserving mitochondrial function during stress conditions .

Research approaches should include comparative analysis of wild-type and HSPB2-knockout models in ischemia-reperfusion injury studies, mitochondrial function assays, and proteomics analysis to identify HSPB2-interacting partners during cardiac stress.

What are the most effective experimental models and designs for studying HSPB2 functions in different cellular contexts?

For studying HSPB2 functions, researchers should consider a multi-tiered approach using complementary models:

  • In vitro cellular models:

    • Primary cultures: Cardiac myocytes and skeletal muscle cells are ideal for studying tissue-specific functions of HSPB2 .

    • Neuronal cultures: Primary neuron cultures subjected to oxygen-glucose deprivation and reperfusion (OGD/r) effectively mimic the hypoxic environment following TBI or stroke .

    • C2C12 mouse myoblast cell line: Useful for studying HSPB2's role during muscle differentiation .

  • In vivo models:

    • Transgenic mouse models: Tamoxifen-induced neuron-specific HSPB2 overexpression models allow temporal control over HSPB2 expression .

    • Knockout models: HSPB2-deficient mice for loss-of-function studies.

    • Disease models: TBI mouse models, cardiac ischemia-reperfusion models, and myotonic dystrophy models.

  • Experimental designs:

    • Comparative analyses between wild-type and HSPB2-modified models.

    • Time-course studies to capture dynamic changes in HSPB2 expression and localization.

    • Controlled stress conditions (e.g., hypoxia, oxidative stress) to evaluate HSPB2's protective functions.

    • Co-expression studies with HSPB3 to investigate heteromeric complex formation and function.

What techniques are most appropriate for analyzing HSPB2 structure and its interactions with binding partners?

To effectively analyze HSPB2 structure and interactions, researchers should employ multiple complementary techniques:

  • Structural analysis techniques:

    • X-ray crystallography: Has been successfully used to determine the structure of human HspB2/B3 heterotetramer .

    • Nuclear Magnetic Resonance (NMR) spectroscopy: Useful for analyzing dynamic regions of HSPB2.

    • Cryo-electron microscopy: Particularly valuable for studying larger HSPB2-containing complexes.

    • Hydrogen-deuterium exchange mass spectrometry: Helps identify flexible regions and binding interfaces.

  • Interaction analysis methods:

    • Co-immunoprecipitation: To identify physiological binding partners.

    • Yeast two-hybrid screening: For discovering novel interactions.

    • Surface plasmon resonance: For determining binding kinetics and affinities.

    • FRET/BRET assays: For studying interactions in living cells.

    • Crosslinking mass spectrometry: To map specific interaction sites.

  • Functional interaction assays:

    • Kinase activity assays: For studying HSPB2's effect on DMPK.

    • Chaperone activity assays: Using aggregation-prone proteins like α-synuclein as substrates .

    • Subcellular fractionation: To study HSPB2 translocation between cellular compartments during stress .

What methodologies are most effective for investigating HSPB2's role in autophagy and neural regeneration?

Based on current research findings, the following methodologies are recommended for investigating HSPB2's role in autophagy and neural regeneration:

  • Autophagy assessment techniques:

    • Autophagic flux assays: Using LC3-II/I ratio measurements with and without lysosomal inhibitors.

    • Fluorescent reporter systems: LC3-GFP or tandem mRFP-GFP-LC3 to distinguish between autophagosomes and autolysosomes.

    • Transmission electron microscopy: To visualize autophagic structures at the ultrastructural level.

    • Western blotting for key autophagy markers: p62, Beclin-1, ATG proteins, and ULK1.

  • Neural regeneration assessment:

    • Behavioral testing battery: Including body curl test, rotarod test, grid-walking test, and cylinder test to assess sensorimotor recovery .

    • Electrophysiological measurements: Callosal compound action potential (CAP) recordings to evaluate white matter functional integrity .

    • Immunohistochemistry: For synaptophysin to assess synaptic density in peri-injury areas .

    • Diffusion tensor imaging: To evaluate white matter structural integrity.

    • Anterograde and retrograde tracing: To assess neural connectivity and remodeling.

  • Gene manipulation approaches:

    • Conditional transgenic models: Such as tamoxifen-induced neuron-specific HSPB2 overexpression mice .

    • AAV-mediated gene delivery: For region-specific manipulation of HSPB2 expression.

    • CRISPR-Cas9 genome editing: For generating precise mutations mimicking human disease variants.

How should researchers address the heterogeneity in HSPB2 structure and function when interpreting experimental results?

HSPB2, like other small heat shock proteins, exhibits considerable heterogeneity across multiple spatiotemporal regimes—from fast fluctuations of protein regions to conformational variability, interface plasticity, and polydispersity of interconverting oligomers . This heterogeneity presents significant challenges for data interpretation.

When addressing this heterogeneity, researchers should:

  • Employ multiple structural techniques:

    • Compare results from different structural methods (X-ray crystallography, NMR, cryo-EM).

    • Consider ensemble-based approaches that capture multiple conformational states.

    • Analyze both isolated domains and full-length proteins to understand domain interactions.

  • Account for differential complex formation:

    • Analyze both homomeric HSPB2 assemblies and heteromeric complexes (particularly HSPB2/B3) .

    • Consider the 3:1 stoichiometry of HSPB2/B3 complexes when designing and interpreting experiments.

    • Investigate how complex formation affects function using targeted mutations at interface regions.

  • Consider tissue-specific differences:

    • Compare HSPB2 behavior across relevant tissues (heart, skeletal muscle, neurons).

    • Investigate tissue-specific binding partners that may modulate HSPB2 function.

    • Account for different expression levels across tissues in functional comparisons.

  • Integrate data across functional assays:

    • Correlate structural features with specific functional outcomes.

    • Use statistical approaches that account for heterogeneity, such as mixed-effects models.

    • Consider developmental and stress-induced changes in HSPB2 behavior.

What approaches are recommended for distinguishing between direct and indirect effects of HSPB2 in cellular protection pathways?

Distinguishing between direct and indirect effects of HSPB2 in cellular protection is challenging due to the complex network of interactions and pathways involved. Researchers should consider the following approaches:

  • Temporal resolution studies:

    • Use time-course experiments with high temporal resolution to establish the sequence of events following HSPB2 activation or manipulation.

    • Employ real-time imaging techniques to track HSPB2 localization and activity.

    • Utilize rapid induction systems (e.g., optogenetics) to precisely control HSPB2 expression or activity.

  • Direct interaction verification:

    • Perform in vitro binding assays with purified components to confirm direct interactions.

    • Use proximity ligation assays in situ to verify interactions in cellular contexts.

    • Employ specific mutations that disrupt binding interfaces to establish causality.

  • Pathway dissection approaches:

    • Use specific inhibitors of downstream pathways to block indirect effects.

    • Perform epistasis experiments by manipulating HSPB2 and potential downstream effectors simultaneously.

    • Employ pathway-specific reporters to monitor activation states.

  • Systems biology approaches:

    • Develop computational models that incorporate known interactions and temporal dynamics.

    • Use network analysis to identify key nodes and potential feedback loops.

    • Perform sensitivity analyses to identify the most influential parameters in HSPB2-mediated protection.

  • Comparative analyses across HSPB family members:

    • Compare effects of different HSPB proteins to identify specific vs. general chaperone effects.

    • Use chimeric proteins with domains from different HSPBs to map functional regions.

How can researchers effectively interpret contradictory findings regarding HSPB2's role in different disease contexts?

Contradictory findings regarding HSPB2's roles in different disease contexts are not uncommon due to its diverse functions and tissue-specific effects. To effectively interpret such contradictions, researchers should:

  • Contextualize disease models:

    • Compare acute vs. chronic disease models (e.g., acute TBI vs. progressive neurodegeneration).

    • Consider disease stage-specific effects—HSPB2 may be protective early but harmful later.

    • Evaluate tissue specificity of effects—protective in heart but potentially different in other tissues.

  • Reconcile cellular mechanism differences:

    • Determine if contradictions arise from different cellular mechanisms being studied.

    • Consider that HSPB2 can both facilitate protein degradation and promote refolding depending on context .

    • Evaluate whether HSPB2 is acting as a monomer, homooligomer, or in complex with HSPB3 .

  • Account for methodological differences:

    • Compare findings from in vitro, ex vivo, and in vivo studies systematically.

    • Consider differences in HSPB2 expression levels across studies (overexpression vs. endogenous).

    • Evaluate the impact of different experimental readouts and endpoints.

  • Perform integrative analyses:

    • Use meta-analysis approaches when multiple studies are available.

    • Consider pathway-focused rather than protein-focused interpretations.

    • Develop unified models that can accommodate seemingly contradictory findings.

  • Design decisive experiments:

    • Identify the source of contradiction and design experiments specifically to address it.

    • Use multiple disease models in parallel with identical methodologies.

    • Include time-course and dose-response studies to capture dynamic effects.

What are the known disease-associated mutations in HSPB2 and their functional consequences?

HSPB2 is associated with several diseases, most notably Myotonic Dystrophy and Malignant Fibrous Histiocytoma . While specific mutations in other HSPBs have been well-characterized and linked to cardiomyopathies, myopathies, motor neuropathies, and cataract , detailed information on specific HSPB2 mutations is more limited.

Disease-associated mutations in HSPBs generally encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or generation of truncated proteins . These mutations can affect oligomerization, chaperone activity, and interactions with client proteins.

For HSPB2 specifically, researchers investigating disease-associated mutations should:

  • Compare the functional consequences with better-characterized mutations in other HSPB family members.

  • Examine effects on HSPB2-HSPB3 complex formation, as disruption of this heteromeric assembly could contribute to pathology.

  • Investigate the impact on DMPK binding and activation, particularly in the context of myotonic dystrophy.

  • Assess changes in subcellular localization, especially translocation to myofibrils during stress conditions.

How might HSPB2-targeting therapeutic approaches be developed for neurodegenerative and neuromuscular disorders?

Based on HSPB2's roles in neuroprotection, muscle maintenance, and stress response, several therapeutic approaches could be developed:

  • Gene therapy approaches:

    • AAV-mediated HSPB2 delivery to neurons for neuroprotection in TBI or neurodegenerative diseases .

    • Muscle-targeted gene therapy for myotonic dystrophy and other muscle disorders.

    • Conditional expression systems to provide temporal control of HSPB2 levels.

  • Small molecule modulators:

    • Compounds that enhance HSPB2 chaperone activity.

    • Stabilizers of HSPB2-HSPB3 heterocomplexes.

    • Molecules that promote HSPB2-mediated autophagy for clearing protein aggregates.

  • Peptide-based therapeutics:

    • Peptides derived from HSPB2 functional domains that can penetrate cells.

    • Stapled peptides that mimic HSPB2-client interactions.

    • Peptide inhibitors of pathological interactions.

  • Combination therapies:

    • HSPB2-targeting approaches combined with other proteostasis modulators.

    • Sequential therapies targeting different stages of disease progression.

    • Tissue-specific targeting strategies to maximize efficacy and minimize off-target effects.

  • Biomarker development:

    • HSPB2 levels or post-translational modifications as prognostic or therapeutic response biomarkers.

    • Monitoring HSPB2 subcellular localization as an indicator of cellular stress.

What emerging technologies and approaches might advance our understanding of HSPB2 biology?

Several cutting-edge technologies hold promise for advancing HSPB2 research:

  • Single-molecule techniques:

    • Single-molecule FRET to study conformational dynamics.

    • Optical tweezers to measure chaperone-substrate interactions at the single-molecule level.

    • Super-resolution microscopy to visualize HSPB2 dynamics in living cells.

  • Advanced structural biology approaches:

    • AlphaFold2 and other AI-based structure prediction tools to model full-length HSPB2 and its complexes.

    • Integrative structural biology combining multiple data sources (X-ray, NMR, cryo-EM, crosslinking MS).

    • Time-resolved structural studies to capture dynamic states.

  • Spatial transcriptomics and proteomics:

    • Spatial mapping of HSPB2 expression and interactions within tissues.

    • Single-cell proteomics to capture cell-specific HSPB2 functions.

    • Proximity labeling approaches to identify context-specific interaction networks.

  • In vivo functional imaging:

    • Genetically encoded sensors for monitoring HSPB2 activity in living animals.

    • Intravital microscopy to observe HSPB2 dynamics during stress responses.

    • PET/MRI imaging with HSPB2-targeted probes for clinical translation.

  • High-throughput functional genomics:

    • CRISPR screens to identify genetic modifiers of HSPB2 function.

    • Systematic mutagenesis to create comprehensive mutation-function maps.

    • Pooled in vivo screens to identify context-specific functions.

What are the most critical unanswered questions regarding HSPB2 function and regulation?

Despite progress in HSPB2 research, several critical questions remain unanswered:

  • Structural dynamics:

    • How do the disordered regions of HSPB2 contribute to its function?

    • What is the structural basis for the specificity of HSPB2-HSPB3 heterocomplex formation?

    • How does HSPB2 recognize and bind to its diverse client proteins?

  • Regulatory mechanisms:

    • What signaling pathways regulate HSPB2 expression and activity?

    • How are HSPB2 post-translational modifications regulated and what are their functional consequences?

    • What determines the balance between HSPB2 homomeric and heteromeric assemblies?

  • Tissue-specific functions:

    • Why is HSPB2 preferentially expressed in heart and skeletal muscle?

    • What are the unique roles of HSPB2 in neurons and how do they differ from its functions in muscle?

    • How does HSPB2 contribute to tissue-specific disease mechanisms?

  • Therapeutic potential:

    • Can HSPB2 modulation provide therapeutic benefits for neurodegenerative diseases beyond TBI?

    • What is the therapeutic window for HSPB2 intervention in acute vs. chronic conditions?

    • How might HSPB2-targeting approaches be personalized based on disease mechanisms?

  • Evolutionary aspects:

    • How has HSPB2 function evolved across species?

    • What are the functional consequences of HSPB2 specialization compared to more conserved heat shock proteins?

Addressing these questions will require interdisciplinary approaches combining structural biology, cell biology, genetics, and systems biology in relevant physiological and pathological contexts.

Product Science Overview

Introduction

Heat Shock 27kDa Protein 2 (HSP27), also known as HSPB1, is a member of the small heat shock protein (sHSP) family. These proteins are highly conserved across species and play crucial roles in cellular protection against stress. HSP27 is particularly notable for its involvement in protein folding, protection against apoptosis, and regulation of the cytoskeleton.

Discovery and Evolution

HSP27 was initially characterized in response to heat shock, a condition where cells are exposed to elevated temperatures. The protein was found to act as a molecular chaperone, facilitating the proper refolding of damaged proteins . Over time, research revealed that HSP27 responds to various stress conditions, including oxidative and chemical stress .

Structure

HSP27 belongs to the small molecular weight heat shock protein family, which ranges from 12 to 43 kDa . It shares a conserved C-terminal domain known as the α-crystallin domain, which is also found in the vertebrate eye lens α-crystallin . This domain is essential for the protein’s oligomerization, a process crucial for its function .

Functions

HSP27 is a multifunctional protein with several roles:

  • Protein Chaperone: It assists in the proper folding of nascent and stress-denatured proteins, preventing their aggregation .
  • Antioxidant: During oxidative stress, HSP27 lowers reactive oxygen species (ROS) levels by increasing intracellular glutathione and reducing intracellular iron .
  • Anti-apoptotic: HSP27 inhibits apoptosis by interacting with both mitochondrial-dependent and independent pathways . It binds to DAXX during Fas-FasL mediated apoptosis, preventing the binding of Ask1 by DAXX . It also interacts with Bax and cytochrome c, preventing mitochondrial-dependent apoptosis .
  • Cytoskeletal Regulation: HSP27 regulates actin cytoskeletal dynamics, promoting actin polymerization and functioning as an actin capping protein .
Clinical Significance

HSP27 has been implicated in various disease states, playing both protective and counter-protective roles. It is involved in renal injury, fibrosis, cancer, neurodegenerative diseases, and cardiovascular diseases . Its anti-apoptotic properties have significant implications for the success of certain chemotherapies .

Recombinant HSP27

Recombinant HSP27 is produced using genetic engineering techniques, where the HSP27 gene is inserted into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the protein, which can be used for research and therapeutic purposes.

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.