HSPA6 Human

Heat Shock 70kDa Protein 6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to HSPA6

HSPA6 (Heat Shock Protein Family A Member 6), also known as HSP70B', is a stress-inducible member of the HSP70 chaperone family. Located on human chromosome 1q23.3, it encodes a protein with two critical domains: an N-terminal nucleotide-binding domain and a C-terminal substrate-binding domain . Unlike other HSP70 isoforms, HSPA6 is minimally expressed under normal conditions but strongly induced during cellular stress, infections, or malignant transformation .

Role in Viral Infections

HSPA6 is exploited by viruses to enhance replication:

  • EV-A71 (Enterovirus A71): HSPA6 induction peaks at 6 hours post-infection, promoting viral RNA translation. Knockout reduces viral protein synthesis by 40–60% and impairs IRES activity .

  • Broad-spectrum IRES activation: Facilitates translation for CV-A16, echovirus 9, and HCV, suggesting a universal role in viral IRES machinery .

Mechanism:

  • Acts on cellular factors (e.g., ITAFs) rather than viral proteins to enhance IRES activity .

  • Does not affect viral assembly or release .

Implications in Cancer

HSPA6 exhibits dual roles in oncology, acting as either an oncogene or tumor suppressor depending on cancer type:

Cancer TypeRole of HSPA6MechanismClinical Impact
GliomaOncogenicPromotes proliferation, invasion, and anti-apoptosis via immune modulation and angiogenesis pathways High expression correlates with poor survival (AUC for 5-year OS: 0.747)
Bladder CancerTumor-suppressiveInhibits tumorigenesis; low expression linked to progression Potential prognostic marker
Lung CancerTumor-suppressiveDownregulation associated with advanced stages Predictive biomarker for therapy

Immune Regulation and Tumor Microenvironment

In gliomas, HSPA6 correlates with immunosuppressive features:

  • Immune infiltration: High HSPA6 expression associates with elevated immune checkpoint molecules (PD-L1, CTLA-4) and immunosuppressive cells (Tregs, M2 macrophages) .

  • Pathway activation: Enriches pathways like JAK-STAT, NF-κB, and antigen presentation .

Key findings:

  • HSPA6⁺ gliomas show higher stromal/immune scores but lower tumor purity .

  • HSPA6 knockdown reduces glioma cell migration by 50% and invasion by 65% in vitro .

Expression in Brain Tissues

Normal vs. diseased states:

  • Normal brain: Low baseline expression .

  • Glioma tissues: Overexpressed compared to adjacent normal tissue (validated via Western blot) .

Regional specificity:

  • Highest expression in cerebral cortex and hippocampus .

Interaction Network

HSPA6 collaborates with co-chaperones and signaling proteins:

InteractorFunctionInteraction Score
DNAJB1Co-chaperone for HSP70; stimulates ATP hydrolysis0.984
HSPA1BChaperone with overlapping stress-response roles0.959
MAPK8/9Regulates apoptosis and stress signaling0.911–0.906

Therapeutic Potential

  • Biomarker: High HSPA6 expression predicts poor prognosis in gliomas (HR = 2.34, p < 0.001) .

  • Target: siRNA-mediated knockdown reduces glioma cell viability by 40% .

  • Challenges: Context-dependent roles necessitate cancer-specific therapeutic strategies .

Product Specs

Introduction
HSPA6, a unique member of the human Hsp70 chaperone family, plays a crucial role in cellular processes such as protein folding, trafficking, and preventing aggregation. Unlike other chaperones, HSPA6 expression is primarily induced by stress, exhibiting minimal to no basal levels in most cells. Both HSPA6 and HSP72 are vital for cell survival under proteotoxic stress conditions.
Description
Recombinant Human HSPA6, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 663 amino acids (specifically, amino acids 1 to 643) with a molecular weight of 73.2 kDa. This recombinant HSPA6 protein is engineered with a 20 amino acid His tag fused at the N-terminus to facilitate purification using standard chromatographic methods.
Physical Appearance
Clear, colorless solution that has undergone sterile filtration.
Formulation
The HSPA6 protein is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0, 100mM sodium chloride (NaCl), and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Minimize repeated freezing and thawing cycles.
Purity
Purity exceeds 90% as assessed by SDS-PAGE analysis.
Synonyms
HSP70B, Heat shock 70 kDa protein 6, Heat shock 70 kDa protein B', HSPA6.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MQAPRELAVG IDLGTTYSCV GVFQQGRVEI LANDQGNRTT PSYVAFTDTE RLVGDAAKSQ AALNPHNTVF DAKRLIGRKF ADTTVQSDMK HWPFRVVSEG GKPKVRVCYR GEDKTFYPEE ISSMVLSKMK ETAEAYLGQP VKHAVITVPA YFNDSQRQAT KDAGAIAGLN VLRIINEPTA AAIAYGLDRR GAGERNVLIF DLGGGTFDVS VLSIDAGVFE VKATAGDTHL GGEDFDNRLV NHFMEEFRRK HGKDLSGNKR ALRRLRTACE RAKRTLSSST QATLEIDSLF EGVDFYTSIT RARFEELCSD LFRSTLEPVE KALRDAKLDK AQIHDVVLVG GSTRIPKVQK LLQDFFNGKE LNKSINPDEA VAYGAAVQAA VLMGDKCEKV QDLLLLDVAP LSLGLETAGG VMTTLIQRNA TIPTKQTQTF
TTYSDNQPGV FIQVYEGERA MTKDNNLLGR FELSGIPPAP RGVPQIEVTF DIDANGILSV TATDRSTGKA NKITITNDKG RLSKEEVERM VHEAEQYKAE DEAQRDRVAA KNSLEAHVFH VKGSLQEESL RDKIPEEDRR KMQDKCREVL AWLEHNQLAE KEEYEHQKRE LEQICRPIFS RLYGGPGVPG GSSCGTQARQ GDPSTGPIIE EVD.

Q&A

What is HSPA6 and how does it differ from other HSP70 family members?

HSPA6 (Heat Shock Protein A6) is a 70-kDa stress-induced heat shock protein cytogenetically located on human chromosome 1q23.3, first identified in 1990 . Unlike constitutively expressed HSP70 family members (HSPA1, HSPA8, and HSPA9) which regulate multiple phases of viral life cycles, HSPA6 has more specialized functions . For example, in viral infections, HSPA6 is specifically required for IRES-mediated translation rather than affecting all phases of the viral life cycle . HSPA6 shares over 90% nucleotide identity with HSPA7 through coding regions, though HSPA7 lacks protein-coding potential .

How is HSPA6 expression regulated in human cells?

HSPA6 expression is primarily regulated through stress-induced pathways. Research indicates complex regulatory mechanisms involving both transcriptional control and epigenetic modifications. In breast cancer studies, researchers found that promoter methylation of HSPA6 is positively correlated with its expression , which differs from the typical inverse relationship between promoter methylation and gene expression. The methylation status of HSPA6 promoter regions shows differences between normal tissues and cancer tissues, suggesting tissue-specific regulatory mechanisms . Additionally, HSPA6 can be induced by specific compounds like thymoquinone (TQ), as demonstrated in triple-negative breast cancer cell lines .

What are the principal methodological approaches to study HSPA6 induction?

To study HSPA6 induction, researchers should employ multiple complementary techniques:

  • Gene expression analysis:

    • RNA-sequencing for genome-wide expression profiling

    • RT-PCR (both standard and quantitative) for targeted validation

    • Western blotting for protein-level confirmation

  • Induction protocols:

    • Heat shock treatment (typically 42-45°C)

    • Chemical inducers (e.g., thymoquinone)

    • Viral infection models (e.g., Enterovirus A71)

  • Functional validation:

    • Overexpression using plasmid transfection

    • Knockdown approaches using shRNA or siRNA

    • Real-Time Cell Analysis (RTCA) to monitor cellular effects

For accurate results, researchers should include appropriate controls and time-course analyses to capture the dynamic nature of HSPA6 induction.

How does HSPA6 contribute to viral infection mechanisms?

HSPA6 functions as a positive regulator during viral infections, particularly in Enterovirus A71 (EV-A71) infection . Research shows that:

  • HSPA6 is specifically required for internal ribosomal entry site (IRES)-mediated translation during viral infection

  • Depletion of HSPA6 leads to reductions in viral proteins, viral RNA, and virions by downregulating IRES-mediated translation

  • Unlike other HSP70 family members that regulate all phases of viral life cycles, HSPA6's role is specialized for the IRES-mediated translation phase

  • HSPA6 facilitates IRES activity through cellular factors rather than direct interaction with viral proteins

  • The importance of HSPA6 extends beyond EV-A71, as knockdown of HSPA6 also reduces luciferase activity driven by the IRES from coxsackievirus A16, echovirus 9, encephalomyocarditis virus, and hepatitis C virus

These findings suggest HSPA6 may assist the function of cellular proteins generally required for viral IRES activities across multiple viral infections.

What are the contradictory roles of HSPA6 in different cancer types?

HSPA6 exhibits context-dependent roles across different cancer types, showing striking contrasts in its effects:

Tumor-promoting roles:

  • In gliomas, HSPA6 enhances malignant progression by promoting proliferation, invasion, and anti-apoptosis

  • High HSPA6 expression correlates with poor clinical prognosis in glioma patients

  • HSPA6 is closely associated with immunity, invasion, and angiogenesis pathways in gliomas

Tumor-suppressive roles:

HSPA6 may also have inhibitory effects in bladder cancer and lung cancer, while being relevant to early recurrence in hepatocellular carcinoma .

How does HSPA6 interact with the tumor microenvironment?

Research indicates significant interactions between HSPA6 and the tumor microenvironment:

  • Immune cell interactions:

    • HSPA6 expression is closely correlated with tumor-infiltrating immune cells (TIICs) in gliomas

    • Different immune checkpoint (ICP) expression levels are observed between patients with low versus high HSPA6 expression

  • Genomic variations:

    • Significant differences in genomic variations exist between tumors with low versus high HSPA6 expression

    • These variations may influence how HSPA6 interacts with the surrounding microenvironment

  • Angiogenesis connections:

    • Gene enrichment analyses show HSPA6 is associated with angiogenesis pathways in gliomas

    • This suggests HSPA6 may influence tumor vasculature development

  • Experimental validation:

    • Comprehensive analyses confirmed significant differences in tumor microenvironments between patients with varying HSPA6 expression levels

These interactions likely contribute to the context-dependent effects of HSPA6 observed across different cancer types.

What are the optimal cell models for studying HSPA6 function?

When selecting cell models for HSPA6 research, researchers should consider:

  • Endogenous expression levels:

    • BT-549 cells: High endogenous HSPA6 expression, suitable for knockdown studies

    • MDA-MB-231 cells: TNBC cell line used to confirm HSPA6 protein expression

    • HeLa cells: Undetectable endogenous HSPA6, ideal for overexpression studies

  • Disease-specific models:

    • For viral studies: Cell lines permissive to viral infection (used for EV-A71, coxsackievirus A16, etc.)

    • For cancer studies: Cell lines representing specific cancer types (glioma cells for brain tumor studies, breast cancer cell lines for TNBC studies)

  • Manipulation approaches:

    • Transfection with HSPA6 plasmids with tags (e.g., Flag) for overexpression

    • shRNA plasmids for successful silencing of endogenous HSPA6

    • CRISPR-Cas9 for complete knockout studies

  • Functional readouts:

    • Real-Time Cell Analysis (RTCA) for monitoring growth, migration, and invasion

    • Cell cycle analysis to assess effects on cell division

    • Virus-specific assays for viral studies (viral RNA quantification, virion production)

How should researchers design experiments to investigate HSPA6's role in IRES-mediated translation?

To effectively study HSPA6's role in IRES-mediated translation, researchers should implement:

  • Reporter systems:

    • Bicistronic reporter constructs containing viral IRES elements

    • Luciferase assays to quantify IRES activity under various conditions

    • Controls comparing cap-dependent and IRES-dependent translation

  • HSPA6 manipulation:

    • Knockdown of HSPA6 using siRNA or shRNA

    • Overexpression of wild-type and mutant HSPA6

    • Time-course analyses to capture dynamic effects

  • Mechanistic investigations:

    • RNA-protein binding assays to assess direct interactions

    • Co-immunoprecipitation to identify relevant protein complexes

    • Domain mutation studies to identify functional regions of HSPA6

  • Validation across viral systems:

    • Test multiple viral IRES elements (EV-A71, coxsackievirus A16, hepatitis C virus)

    • Include non-viral IRES elements as controls

    • Assess effects in different cell types to determine universality

Research has shown that HSPA6 facilitates IRES activity even in the absence of viral proteins, suggesting it works through cellular factors rather than direct viral interactions .

What statistical approaches are most appropriate for analyzing HSPA6 expression in clinical datasets?

For robust statistical analysis of HSPA6 in clinical data, researchers should employ:

  • Survival analysis methods:

    • Kaplan-Meier survival plots with appropriate patient stratification

    • Cox proportional hazards regression for multivariable analysis

    • Time-dependent ROC curves and AUC values to evaluate predictive accuracy

  • Expression correlation analyses:

    • Pearson and Spearman correlations for examining relationships between HSPA6 expression and other variables

    • Methylation-expression correlation to understand epigenetic regulation

  • Multiple cohort validation:

    • Analysis across independent datasets (TCGA, CGGA, GSE16011)

    • Consistent methodology across cohorts

    • Meta-analysis approaches to integrate findings

  • Clinical variable consideration:

    • Stratification by relevant factors (IDH mutations, 1p19q co-deletion, grade, gender, age)

    • Multivariate models to control for confounding variables

    • Subgroup analyses (e.g., specific cancer subtypes)

The research demonstrates that these approaches revealed significant correlations between HSPA6 expression and survival outcomes in both glioma (negative correlation) and breast cancer patients (positive correlation) .

How does HSPA6 regulate cellular signaling pathways differently across tissue types?

The tissue-specific effects of HSPA6 likely result from differential regulation of signaling pathways:

  • In gliomas:

    • GO-BP analysis, KEGG enrichment, and GSVA indicate HSPA6 primarily correlates with immunity, invasion, and angiogenesis pathways

    • Experimental validation confirms HSPA6's role in promoting malignant progression through proliferation, invasion, and anti-apoptosis mechanisms

  • In breast cancer:

    • HSPA6 shows inhibitory effects on growth, migration, and invasion in TNBC cells

    • The mechanism appears distinct from its role in gliomas, suggesting tissue-specific pathway engagement

  • Potential mechanisms for context-dependent effects:

    • Tissue-specific protein interaction networks

    • Differential post-translational modifications

    • Varying subcellular localization

    • Distinct epigenetic regulation

  • Methodological approaches to investigate pathway differences:

    • Pathway analysis tools (GO-BP, KEGG, GSVA)

    • Protein-protein interaction (PPI) network construction

    • Differential gene expression analysis using RNA-seq data

What molecular mechanisms explain HSPA6's role in cancer cell invasion and migration?

HSPA6's effects on invasion and migration involve complex molecular mechanisms:

  • Experimental evidence:

    • In gliomas, HSPA6 promotes invasion

    • In TNBC, HSPA6 inhibits migration and invasion

    • Real-Time Cell Analysis (RTCA) assays confirm these effects

  • Pathway involvement:

    • KEGG pathway analysis reveals HSPA6 correlates with invasion-related pathways in gliomas

    • The contrasting effects in different cancers suggest tissue-specific mechanisms

  • Potential molecular mechanisms:

    • Regulation of cytoskeletal remodeling proteins

    • Effects on cell adhesion molecules

    • Modulation of extracellular matrix degradation enzymes

    • Influence on epithelial-mesenchymal transition (EMT) processes

  • Research approaches to dissect these mechanisms:

    • Analysis of invasion-related gene expression following HSPA6 manipulation

    • Imaging studies to visualize cytoskeletal changes

    • Matrix degradation assays

    • In vivo metastasis models

How does HSPA6 interact with other HSP family members in stress response and disease?

While research on HSPA6's interactions with other HSP family members is limited, several insights can be drawn:

  • Differential roles compared to other HSP70 proteins:

    • Unlike HSPA1, HSPA8, and HSPA9 which regulate all phases of viral life cycles, HSPA6 is specifically required for IRES-mediated translation

    • This functional specialization suggests distinct interactions with cofactors and client proteins

  • Co-expression relationships:

    • Network analysis identified nine heat shock proteins differentially expressed in gliomas, with HSPA6 being the only one from the HSP70 family

    • This suggests potential coordinated regulation within the broader heat shock response

  • Methodological approaches to study interactions:

    • Protein-protein interaction networks through computational prediction and experimental validation

    • Co-immunoprecipitation to identify physical interactions

    • Co-expression analysis across different stress conditions

    • Functional redundancy studies through combinatorial knockdown experiments

  • Therapeutic implications:

    • Understanding the interaction profile could inform more selective targeting strategies

    • Combination approaches targeting multiple HSP family members might be more effective in certain contexts

How might HSPA6 be targeted therapeutically in cancer treatment?

Therapeutic targeting of HSPA6 would require context-specific approaches based on its divergent roles:

  • For cancers where HSPA6 promotes malignancy (e.g., gliomas):

    • Small molecule inhibitors targeting HSPA6's ATPase activity

    • siRNA or antisense oligonucleotides for expression knockdown

    • Disruption of key protein-protein interactions

    • HSPA6 serves as a promising therapeutic target to improve the prognosis of glioma patients

  • For cancers where HSPA6 suppresses malignancy (e.g., breast cancer):

    • Approaches to upregulate or stabilize HSPA6 expression

    • Protection of HSPA6 from degradation

    • Activation of pathways that induce HSPA6 expression

  • Considerations for therapeutic development:

    • Specificity over other HSP70 family members

    • Tissue-specific delivery systems

    • Potential combination with standard therapies

    • Biomarker-guided patient selection

Research validating HSPA6 as a therapeutic target requires further preclinical studies and careful consideration of context-dependent effects.

What is the potential for HSPA6 as a prognostic biomarker in cancer?

HSPA6 shows significant potential as a prognostic biomarker:

The opposing prognostic associations in different cancers highlight the importance of context-specific biomarker validation.

How can researchers translate findings about HSPA6 in viral infections to antiviral therapeutic development?

Translating HSPA6 research to antiviral therapeutics involves several strategic approaches:

  • Targeting HSPA6-mediated IRES translation:

    • HSPA6 is required for IRES-mediated translation in multiple viruses (EV-A71, coxsackievirus A16, echovirus 9, encephalomyocarditis virus, hepatitis C virus)

    • Inhibiting HSPA6 could potentially block viral protein synthesis

  • Mechanistic understanding for drug development:

    • HSPA6 facilitates IRES activity through cellular factors rather than direct viral interaction

    • Identifying these cellular factors could provide additional therapeutic targets

    • Targeting the HSPA6-cellular factor interaction might offer more specificity than directly targeting HSPA6

  • Experimental approaches for therapeutic development:

    • High-throughput screening for HSPA6 inhibitors

    • Structure-based drug design targeting HSPA6's functional domains

    • Peptide inhibitors to disrupt specific protein-protein interactions

    • Testing candidates in viral infection models measuring viral proteins, RNA, and virion production

  • Considerations for antiviral applications:

    • Spectrum of activity across different viruses utilizing IRES-mediated translation

    • Potential off-target effects on cellular IRES-containing mRNAs

    • Combination approaches with established antivirals

What are the most pressing questions that remain unanswered about HSPA6 function?

Several critical knowledge gaps in HSPA6 biology require further investigation:

  • Molecular mechanisms underlying context-dependent effects:

    • What explains HSPA6's tumor-promoting role in gliomas versus tumor-suppressive role in breast cancer?

    • What tissue-specific interaction partners mediate these divergent functions?

  • Detailed structure-function relationships:

    • Which domains of HSPA6 are responsible for its specialized functions?

    • How does HSPA6's structure differ from other HSP70 family members?

  • Regulation of HSPA6 expression:

    • What factors control the stress-induced expression of HSPA6?

    • Why is promoter methylation positively correlated with HSPA6 expression in breast cancer?

  • Role in normal physiology:

    • What is HSPA6's function in non-pathological conditions?

    • Why is HSPA6 stress-inducible rather than constitutively expressed?

  • Therapeutic targeting:

    • How can HSPA6 be selectively targeted without affecting other HSP70 family members?

    • What biomarkers can predict response to HSPA6-targeted therapies?

What emerging technologies could accelerate HSPA6 research?

Advanced technologies that could drive HSPA6 research forward include:

  • Single-cell approaches:

    • Single-cell RNA-seq to capture heterogeneity in HSPA6 expression

    • Single-cell proteomics to examine protein-level dynamics

    • Spatial transcriptomics to map HSPA6 expression within tissue context

  • CRISPR-based technologies:

    • CRISPR screening to identify synthetic lethal interactions

    • CRISPRi/CRISPRa for precise modulation of HSPA6 expression

    • Base editing for introducing specific mutations in endogenous HSPA6

  • Structural biology methods:

    • Cryo-EM to determine HSPA6's structure and conformational states

    • Hydrogen-deuterium exchange mass spectrometry to study dynamics

    • Proximity labeling (BioID, APEX) to map interaction networks

  • Computational approaches:

    • Machine learning to predict HSPA6 interactions and functions

    • Systems biology modeling of HSPA6 in stress response networks

    • Virtual screening for HSPA6-targeting compounds

  • Organoid and advanced cell culture systems:

    • Patient-derived organoids to study HSPA6 in physiologically relevant models

    • Microfluidic systems to analyze HSPA6 dynamics under controlled stress conditions

How might our understanding of HSPA6 inform research on other stress-response proteins?

Insights from HSPA6 research provide valuable paradigms for studying stress-response proteins:

  • Context-dependent functions:

    • HSPA6's varying roles across tissues illustrates how stress proteins may have specialized functions beyond canonical chaperone activity

    • This suggests other stress proteins may similarly show tissue-specific effects

  • Methodological approaches:

    • Multi-omics integration strategies used for HSPA6 research

    • Experimental designs comparing effects across different cell types and disease contexts

    • Bioinformatic approaches correlating expression with clinical outcomes

  • Therapeutic implications:

    • The dual nature of HSPA6 in different cancers highlights the importance of context-specific targeting

    • Potential for stress proteins as both biomarkers and therapeutic targets

  • Evolutionary considerations:

    • HSPA6 shows distinctive features compared to other HSP70 family members

    • Comparative studies may reveal how stress-response proteins evolve specialized functions

  • Systems biology perspective:

    • HSPA6 research demonstrates how a single stress protein can impact multiple cellular pathways

    • This supports network-based approaches to understanding stress-response systems

Product Science Overview

Introduction

Heat Shock 70kDa Protein 6 (HSPA6) is a member of the Heat Shock Protein 70 (HSP70) family, which is a group of highly conserved proteins that play a crucial role in cellular processes such as protein folding, trafficking, and prevention of aggregation . These proteins are known for their ability to protect cells from stress-induced damage by acting as molecular chaperones.

Discovery and Function

The concept of heat shock proteins was first discovered in the 1960s by Ferruccio Ritossa, who observed an unusual “puffing pattern” in the chromosomes of Drosophila (fruit flies) when exposed to elevated temperatures . This led to the identification of the Heat Shock Response and the subsequent discovery of Heat Shock Proteins (HSPs).

HSPA6, specifically, is a stress-inducible protein, meaning it is produced in response to cellular stressors such as heat, heavy metals, and other toxic chemicals . Unlike some other members of the HSP70 family, HSPA6 has little to no basal expression in most cells under normal conditions .

Structure

HSPA6, like other HSP70 proteins, consists of three major functional domains:

  1. N-terminal ATPase domain: This domain binds and hydrolyzes ATP (Adenosine triphosphate) to ADP (Adenosine diphosphate), driving conformational changes necessary for its chaperone activity.
  2. Substrate-binding domain (SBD): This domain interacts with extended polypeptides, helping in the proper folding of nascent proteins and the refolding of misfolded proteins.
  3. C-terminal domain: This domain contains an EEVD motif that is involved in binding to co-chaperones and other HSPs .
Industrial Production

Recombinant HSPA6 is typically produced using bacterial or mammalian expression systems. The gene encoding HSPA6 is cloned into an expression vector, which is then introduced into host cells such as Escherichia coli or HEK293 cells. The recombinant protein is expressed, harvested, and purified using techniques such as affinity chromatography .

Applications

HSPA6 has significant potential in various biomedical applications. It is used in research to study cellular stress responses and protein folding mechanisms. Additionally, due to its role in protecting cells from stress-induced damage, HSPA6 is being explored for therapeutic applications, including cancer treatment and neurodegenerative diseases .

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.