STX6 Human

Syntaxin-6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Protein Structure and Domains

The STX6 protein contains several functional domains that are critical for its activity in vesicular trafficking:

1.1.1 N-terminal Domain

The N-terminal domain of STX6 adopts a structure consisting of an antiparallel three-helix bundle . Despite low sequence similarity, this domain shows strong structural similarity with the N-terminal domains of other syntaxins, including syntaxin 1a, Sso1p, and Vam3p . The N-terminal domain has regulatory functions, binding to the SNARE motif and decreasing the rate of association with partner SNAREs .

1.1.2 SNARE Motif

The SNARE motif of STX6 shows significant homology to both syntaxin 1a and S25C, indicating evolutionary conservation . This motif is essential for the formation of the four-helix coiled-coil structure that brings vesicle and target membranes together for fusion .

1.1.3 Transmembrane Region

The C-terminal region contains a transmembrane domain that anchors the protein to membranes. This region is characterized by a hydrophobic sequence: WCAIAILFAVLLVVLILFLVL .

Functional Roles of STX6

STX6 serves as a target SNARE (t-SNARE) primarily found in endosomal transport vesicles . Its functions center around facilitating intracellular vesicle trafficking and membrane fusion events.

Vesicular Transport Mechanisms

STX6 functions essentially as a tether to hold vesicles during trafficking events . The cytoplasmic regions of SNARE proteins found on transport vesicles and target membranes interact, forming a four-helix coiled coil that links cell membranes and vesicles together in a manner that overcomes the energetic barrier to fusing two lipid bilayers . This mechanism enables the exchange of cellular cargo between different compartments.

Subcellular Localization

The protein co-localizes with vesicle-associated membrane protein (VAMP) 4 to tubular and vesicular membranes of the Golgi apparatus . Subcellular localization studies have identified STX6 in the cytoplasm and cell membrane . This strategic positioning enables STX6 to participate in multiple trafficking pathways within the cell.

Regulatory Functions

STX6 demonstrates important regulatory roles in:

  1. The delivery of microdomain-associated lipids and proteins to the cell surface

  2. Influencing the reinternalization of glucose transporter 4 (GLUT4) upon insulin withdrawal

  3. Modulating secretion in activated macrophages

Expression Profile of STX6 in Human Tissues

STX6 shows a broad tissue distribution pattern across the human body, with particularly notable expression in specific organs.

Tissue-Specific Expression Patterns

While STX6 is widely expressed throughout the body, higher expression levels have been documented in:

  • Brain

  • Lung

  • Kidney

This expression profile can be visualized through resources like The Human Protein Atlas, which provides data regarding STX6 expression at both mRNA and protein levels .

TissueRelative Expression Level
BrainHigh
LungHigh
KidneyHigh
LiverModerate
Other tissuesVariable

Protein Interactions and Pathways

STX6 functions within complex networks of protein interactions that facilitate its role in cellular trafficking.

Protein-Protein Interactions

Research has demonstrated that STX6 interacts with several proteins involved in vesicular trafficking:

  • SNAP23

  • VAMP3

  • VAMP4

These interactions form the basis of functional SNARE complexes that mediate specific membrane fusion events throughout the cell.

Involvement in Cellular Pathways

STX6 is primarily involved in the SNARE interactions in vesicular transport pathway . Its positioning within this pathway makes it a critical component for maintaining normal cellular homeostasis and responding to changing cellular needs.

Role of STX6 in Disease Pathogenesis

Recent research has implicated STX6 in several pathological processes, highlighting its potential significance in both diagnosis and treatment strategies.

STX6 in Hepatocellular Carcinoma

Immunohistochemistry studies have demonstrated that STX6 protein is highly expressed in hepatocellular carcinoma (HCC) tissues compared to paired adjacent tissues . The protein shows significant associations with several clinicopathological features:

5.1.1 Clinical Associations

STX6 protein expression in HCC is significantly associated with:

  • Tumor size (P=0.003)

  • Edmondson grade (P=0.020)

  • Alpha-fetoprotein (AFP) level (P=0.019)

5.1.2 Immune Infiltration Correlation

STX6 expression demonstrates significant positive correlations with immune cell infiltration in HCC:

Immune Cell TypeCorrelation Coefficient (r)P-value
B cells0.3896.93×10^-14
CD4+ T cells0.5411.50×10^-27
CD8+ T cells0.2473.77×10^-6
Macrophages0.5351.17×10^-26
Neutrophils0.4573.11×10^-19
Dendritic cells0.4161.15×10^-15

5.1.3 Tumor-Associated Macrophage Markers

STX6 expression also shows positive associations with tumor-associated macrophage (TAM) markers:

TAM MarkerCorrelation Coefficient (r)P-value
CCL20.2711.28×10^-7
CD680.2985.38×10^-9
IL100.311.11×10^-9
MS4A4A0.2029.06×10^-5
MSR10.3711.62×10^-13
VSIG40.2221.65×10^-5

These findings suggest that STX6 may play a role in modulating the immune microenvironment in HCC, potentially contributing to tumor progression through immunosuppressive mechanisms.

STX6 in Neurodegenerative Diseases

Recent genetic and experimental evidence has implicated STX6 in several neurodegenerative conditions.

5.2.1 Role in Prion Diseases

Human genetics studies have linked an STX6 gene variant to increased risk of developing sporadic Creutzfeldt-Jakob disease (sCJD) . People with this risk variant have approximately 16% higher chance of developing sCJD and produce higher levels of STX6 protein .

Experimental evidence from cellular models demonstrates that modulation of STX6 levels affects prion susceptibility:

  • Knockdown of STX6 alters the distribution of disease-related prion protein (PrP)

  • STX6 knockdown increases perinuclear accumulation of disease-related PrP

  • STX6 knockdown reduces plasma membrane staining of disease-related PrP

These findings suggest that STX6 influences prion disease pathogenesis through effects on protein trafficking and cellular distribution.

5.2.2 Impact on Tauopathies

Studies using a transgenic P301S tau mouse model crossed with STX6 knockout mice have revealed significant protective effects:

  • STX6 knockout mice showed improved motor performance on rotarod tests

  • STX6 knockout mitigated frailty measures in tauopathy models

  • STX6 knockout reduced neurodegeneration-related pathology

These protective effects occurred despite increased localized tau pathology in some brain regions, suggesting that STX6 influences the distribution and toxicity of pathological tau proteins through altered trafficking mechanisms.

STX6 as a Biomarker and Therapeutic Target

The involvement of STX6 in multiple disease processes suggests potential applications in both diagnostics and therapeutics.

Diagnostic Potential

The elevated expression of STX6 in HCC and its association with clinicopathological features suggest its potential as a biomarker:

  • STX6 expression is associated with poor prognosis in HCC patients

  • The combination of STX6 with AFP may have higher diagnostic value than AFP alone

  • STX6 may serve as a valuable novel tumor marker for HCC diagnosis

Therapeutic Implications

The discovery that modulating STX6 levels affects disease progression in neurodegenerative models highlights potential therapeutic approaches:

  • Targeting STX6-dependent trafficking pathways may offer new strategies for treating prion diseases

  • Modulation of STX6 expression or function could potentially mitigate tau-related neurodegeneration

  • Understanding the mechanism of STX6 in disease may reveal additional therapeutic targets in the trafficking pathway

Research Tools and Resources

Multiple resources are available for studying STX6 in research settings, enabling further investigation of its functions and potential applications.

STX6 antibodies are available for various applications:

ApplicationDilution RangeHost
Western Blot1:300-5000Rabbit
ELISA1:500-1000Rabbit
IHC-P1:200-400Rabbit
Immunofluorescence1:50-200Rabbit

Animal Models

Genetically modified mouse models have been developed to study STX6 function:

  • STX6 knockout mice

  • STX6 overexpression mice

  • These models allow investigation of STX6's role in both normal development and disease processes

Product Specs

Introduction
STX6, a member of the syntaxin family, possesses a single t-SNARE coiled-coil homology domain and participates in intracellular vesicle trafficking. It interacts with various proteins including EEA1, VPS45A, MARCH2, MARCH3, and GOPC. STX6 is also found within a complex composed of STX6, STX12, VAMP4, and VTI1A.
Description
Recombinant human STX6, expressed in E. coli, is a non-glycosylated polypeptide chain with a molecular weight of 29.2 kDa. The protein consists of 257 amino acids, with residues 1-234 corresponding to the STX6 sequence. A 23 amino acid His-tag is fused to the N-terminus to facilitate purification, which is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The STX6 solution is provided at a concentration of 0.5 mg/ml and is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 2 mM DTT, and 50% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product in frozen form at -20°C. The addition of a carrier protein, such as 0.1% HSA or BSA, is advised for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
SDS-PAGE analysis indicates a purity level exceeding 90.0%.
Synonyms
Syntaxin 6.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSMEDPF FVVKGEVQKA VNTAQGLFQR WTELLQDPST ATREEIDWTT NELRNNLRSI EWDLEDLDET ISIVEANPRK FNLDATELSI RKAFITSTRQ VVRDMKDQMS TSSVQALAER KNRQALLGDS GSQNWSTGTT DKYGRLDREL QRANSHFIEE QQAQQQLIVE QQDEQLELVS GSIGVLKNMS QRIGGELEEQ AVMLEDFSHE LESTQSRLDN VMKKLAKVSH MTSDRRQ

Q&A

Basic Research Questions

  • What is STX6 and what is its fundamental function in human cells?

    STX6 (Syntaxin-6) is a protein that regulates the transport of proteins and other molecules within cells. It primarily functions to help transport cargo for recycling or degradation and delivers cellular cargo to different compartments within the cell . As a trafficking protein, STX6 is involved in the vesicular transport system, which is essential for maintaining cellular homeostasis. Its expression is particularly notable in certain brain regions such as the caudate and putamen, where increased expression has been linked to disease states . The protein is encoded by the STX6 gene, which produces mRNA that is then translated into the functional STX6 protein that participates in these critical trafficking pathways .

  • How is STX6 expression measured in human brain tissue samples?

    STX6 expression in human brain tissue can be measured through several methodological approaches:

    • RNA sequencing (RNA-seq): This technique quantifies mRNA levels of STX6 in tissue samples, as evidenced by studies utilizing TCGA database information (tumor: n=374; normal: n=50) .

    • Immunohistochemistry (IHC): IHC scoring systems are used to quantify STX6 protein expression in tissue sections. As demonstrated in hepatocellular carcinoma studies, IHC scores below 6 are typically classified as low expression, while scores of 6 or higher indicate high expression .

    • Expression Quantitative Trait Loci (eQTL) analysis: This approach identifies genetic variants that affect gene expression levels. Research has utilized eQTL databases to determine that certain STX6 variants are associated with increased STX6 expression in disease-relevant brain regions .

    • Western blotting: This technique is employed for semi-quantitative analysis of STX6 protein levels in tissue homogenates.

  • What cellular pathways involve STX6 protein in neurons?

    STX6 participates in several critical neuronal pathways:

    • Endolysosomal system: STX6 plays a key role in trafficking within the endolysosomal system, which is critical for protein degradation and recycling in neurons .

    • Trans-Golgi network trafficking: STX6 regulates the transport of proteins from the trans-Golgi network to various cellular compartments, including endosomes and the plasma membrane.

    • Protein uptake and release: STX6 is important for neurons to take up and release proteins, which may have implications for intercellular communication .

    • Prion-like protein transmission: Research indicates STX6 may be involved in the cell-to-cell transmission of misfolded proteins such as tau and prion proteins, potentially influencing the progression of neurodegenerative diseases .

  • How does STX6 contribute to protein trafficking in neurons compared to other cell types?

    In neurons, STX6 contributes to protein trafficking with several specialized functions:

    • Neurons have unique compartmentalization needs due to their complex morphology (axons, dendrites, and synaptic terminals), making STX6's role in directed protein transport particularly critical.

    • STX6 regulates the trafficking of synaptic vesicles and neurotransmitter receptors, which are essential for neuronal communication.

    • In the context of neurodegenerative diseases, STX6 appears to influence the uptake and transmission of disease-associated proteins like tau and prion proteins between neurons .

    • Research focusing on cultured human neurons specifically manipulates STX6 expression to study tau protein uptake, highlighting its significance in neuronal protein trafficking .

    Unlike non-neuronal cells, where STX6 primarily mediates general vesicular transport, in neurons it has the additional complexity of managing protein transport across vastly greater distances and maintaining the specialized protein composition of different neuronal compartments.

  • What techniques are used to manipulate STX6 expression in experimental research?

    Several methodological approaches are employed to manipulate STX6 expression:

    • RNA silencing: This technique is used in immortalized cell lines to reduce STX6 expression and study its functional consequences .

    • CRISPR-Cas9 gene editing: This "molecular scissors" approach creates precise modifications in the STX6 gene. It has been used to generate:

      • Complete knockout models (STX6-/-) where the gene is deleted

      • Targeted modifications in human iPSC-derived cortical neurons

    • Single-cell embryo microinjection: For creating genetically modified mice, researchers use microinjection techniques to modify mouse embryos, which are then reimplanted for development of STX6 modified mouse lines .

    • Vector-based overexpression: To model increased STX6 expression (mirroring the effect of risk variants), researchers use viral vectors or plasmid constructs to increase STX6 levels in experimental systems .

    These techniques allow researchers to model both loss and gain of STX6 function to understand its biological significance in various disease contexts.

Advanced Research Questions

  • How do genetic variants in STX6 affect neurodegenerative disease risk in humans?

    Genetic variants in and around the STX6 gene have been significantly associated with neurodegenerative disease risk through several mechanisms:

    • Sporadic Creutzfeldt-Jakob disease (sCJD): Genome-wide association studies (GWAS) have identified STX6 as one of only three genes (alongside PRNP and GAL3ST1) that confer risk for sCJD at genome-wide significance in populations with European ancestry .

    • Risk quantification: People with the STX6 risk variant have approximately a 16% higher chance of developing sCJD compared to those without the variant .

    • Expression effects: The risk variants appear to increase STX6 expression in disease-relevant brain regions, particularly the caudate and putamen, suggesting a causal mechanism involving elevated STX6 levels .

    • Progressive Supranuclear Palsy (PSP): STX6 genetic variants have also been associated with PSP risk, with evidence suggesting that the gene's role in protein trafficking may influence tau protein pathology .

    • Functional impact: The variants likely affect the cellular uptake and cell-to-cell transmission of disease-associated proteins like prions and tau, potentially accelerating disease progression through enhanced protein spread mechanisms .

  • What is the role of STX6 in prion protein cell-to-cell transmission?

    STX6 appears to play a significant role in prion protein cell-to-cell transmission through several mechanisms:

    • Prion-like propagation: STX6 is implicated in the prion-like progression of neurodegenerative diseases, where misfolded proteins spread from one cell to another throughout the nervous system .

    • Experimental evidence: Mouse models with genetic depletion of STX6 (STX6-/-) show modest protective effects against prion disease, particularly when infected with very low concentrations of prions. This suggests STX6 may influence how brain cells handle early prion infection .

    • Dose-dependent effects: At high prion doses (1%), removal of STX6 has limited impact, but at lower doses, the effect becomes more pronounced, indicating STX6's role may be most critical during initial stages of infection .

    • Cellular mechanisms: As a protein involved in vesicular trafficking, STX6 likely regulates the endocytosis, processing, and/or release of prion proteins, thereby affecting their ability to propagate between cells.

    • Therapeutic implications: While STX6 lowering does not appear promising for treating established prion disease, understanding its role in early disease propagation may reveal new strategies for preventing initial spread .

  • How does STX6 expression correlate with clinical outcomes in prion diseases?

    The relationship between STX6 expression and clinical outcomes in prion diseases involves several important aspects:

    ParameterLow STX6 ExpressionHigh STX6 ExpressionStatistical Significance
    Disease RiskLower risk of sCJD16% higher risk of sCJDGenome-wide significance
    Disease ProgressionPotentially slower in mouse modelsPotentially acceleratedObserved in experimental models
    Response to Very Low Dose Prion ExposureMore resistantMore susceptibleMost pronounced at low doses
    Impact on Established DiseaseMinimal therapeutic effectNot testedLimited impact on late-stage disease

    The research indicates that:

    • Genetically determined higher STX6 expression correlates with increased susceptibility to developing sporadic CJD .

    • In mouse models, STX6 depletion shows modest protective effects, primarily during early disease stages or with low prion concentrations .

    • The impact appears to be most significant in relation to disease initiation rather than progression once established, suggesting a potential window for intervention early in the disease process .

    • These findings suggest that STX6 expression levels could potentially serve as a prognostic indicator for individuals at risk of prion diseases, particularly when combined with other genetic risk factors.

  • What methodological challenges exist in studying STX6's role in human neurodegeneration?

    Researchers face several significant methodological challenges when investigating STX6's role in human neurodegeneration:

    • Causal relationship determination: Distinguishing whether STX6 alterations are causes or consequences of neurodegeneration requires sophisticated experimental designs and temporal analyses that are difficult to implement in human studies .

    • Cell-type specificity: STX6 may function differently across various neural cell types. Current techniques have limitations in studying cell-type-specific effects in intact human brain tissue .

    • Limitations of animal models: While mouse models provide valuable insights, there are inherent challenges in translating findings to human disease. For example, mouse STX6-/- studies show modest effects that may not fully reflect human pathophysiology .

    • Temporal dynamics: STX6's impact appears more significant in early disease stages, making it challenging to study its role throughout disease progression, particularly in slowly developing human neurodegenerative conditions .

    • Technical barriers: Manipulating STX6 expression in human neurons requires advanced techniques such as CRISPR-mediated gene editing in iPSC-derived neurons, which have technical limitations including variability and maturation issues .

    • Integration of genetic and functional data: Correlating genetic variants identified in GWAS with functional consequences requires complex multi-omics approaches and large sample sizes that are difficult to achieve in human brain studies .

  • How do mouse models of STX6 modification translate to human disease mechanisms?

    The translation between mouse STX6 models and human disease mechanisms involves several important considerations:

    • Genetic homology: Approximately 98% of genes in humans are also present in mice, with most performing similar functions, providing a foundation for translational research .

    • Disease recapitulation: Mouse models of prion disease successfully replicate many hallmarks of human disease in a shorter timeframe, allowing for accelerated study of disease mechanisms .

    Mouse Model TypeKey FindingsHuman Disease Relevance
    STX6 knockout (STX6-/-)Modest protective effect against prion disease, particularly at low prion dosesSuggests human STX6 variants may affect early disease stages
    STX6 overexpressionEngineered to mirror human risk variant effectsModels increased STX6 levels associated with disease risk in humans
    Cell distribution studiesChanges in subcellular localizationConsistent with altered trafficking role in human neurons
    • Translational limitations: High-dose prion infection models (1%) in mice may mask subtle STX6 effects that could be significant in human disease, which typically involves much lower initial prion concentrations .

    • Humanized models: To better bridge the gap, researchers have developed humanized STX6 overexpression mice that more closely model the effects of human STX6 risk variants .

    • Complementary approaches: Combining knockout and overexpression mouse models with human iPSC-derived neuronal cultures provides a more comprehensive understanding of STX6's role across species .

  • How does alteration of STX6 expression affect tau protein pathology in human neurons?

    The relationship between STX6 expression and tau protein pathology in human neurons involves several mechanisms:

    • Cell-to-cell transmission: STX6 appears to regulate the uptake and transmission of tau protein between neurons. Manipulating STX6 expression in cultured human neurons affects how tau protein is taken up, potentially influencing the spread of tau pathology in diseases like Progressive Supranuclear Palsy (PSP) .

    • Vesicular trafficking: As a component of the cellular trafficking machinery, STX6 likely influences the intracellular processing and sorting of tau protein, affecting its propensity to form aggregates and spread between cells .

    • Disease progression: By mediating tau protein transmission, STX6 may influence the progression pattern of tauopathies throughout the nervous system. This could explain why certain genetic variants of STX6 are associated with tauopathies like PSP .

    • Experimental approaches: Researchers manipulate STX6 expression in cultured human neurons and study tau uptake to elucidate these mechanisms. These methodologies provide insight into how STX6 levels might modify tau-related pathological processes in the human brain .

    • Therapeutic implications: Understanding how STX6 affects tau pathology could reveal potential therapeutic targets for halting the progression of tauopathies by interfering with cell-to-cell transmission mechanisms .

  • How does STX6 interact with other genetic risk factors in neurodegenerative diseases?

    STX6 operates within a complex genetic landscape of neurodegenerative disease risk:

    • Multi-locus risk architecture: Genome-wide association studies have identified STX6 as functioning alongside other risk genes such as PRNP (encoding the prion protein) and GAL3ST1 (encoding cerebroside sulfotransferase) in sporadic CJD .

    • Synergistic effects: The combined presence of risk variants in STX6 and other genes likely produces synergistic effects on disease susceptibility that exceed the sum of their individual contributions.

    • Pathway convergence: STX6 and other risk genes may converge on common cellular pathways such as protein trafficking, lysosomal function, and protein quality control. For instance, STX6's role in endolysosomal trafficking may interact with other genes involved in protein degradation pathways .

    • Genetic risk stratification: Understanding the interactions between STX6 and other risk factors could enable more accurate genetic risk profiling for neurodegenerative diseases.

    • Genetic background effects: The impact of STX6 variants may be modified by the broader genetic background of an individual, explaining variable penetrance and expressivity of disease across populations.

    • Therapeutic targeting considerations: The interconnected nature of these genetic risk factors suggests that therapeutic approaches may need to target multiple pathways simultaneously for maximum efficacy.

  • What are the current experimental approaches to target STX6 for therapeutic intervention?

    Several experimental approaches are being explored to target STX6 for potential therapeutic interventions:

    • Expression modulation: As increased STX6 expression appears to elevate disease risk, approaches to normalize or reduce STX6 expression in disease-relevant brain regions are being investigated .

    • Timing considerations: Research indicates that STX6-targeting interventions may be most effective during early disease stages or as preventative measures in high-risk individuals, rather than for treating established disease .

    • Targeted delivery systems: Development of brain-penetrant delivery systems for STX6-modulating compounds or genetic interventions represents an active area of research.

    • Proof-of-concept studies: Mouse models with genetic depletion of STX6 demonstrate modest protective effects against prion disease, providing proof-of-concept for STX6 as a potential therapeutic target, particularly for early intervention .

    • Combination approaches: Given that STX6 is one of multiple genetic risk factors for neurodegenerative diseases, combination therapies targeting several pathways simultaneously may offer more comprehensive protection.

    • Biomarker development: STX6 expression levels and activity might serve as biomarkers for disease susceptibility or progression, potentially guiding personalized therapeutic approaches based on individual risk profiles .

    The current evidence suggests that while STX6 lowering alone may not be sufficient for treating established prion disease, further investigation into its role during early disease stages could yield valuable therapeutic insights .

Product Science Overview

Structure and Localization

Syntaxin-6 is characterized by a single-pass type IV membrane protein structure and contains a t-SNARE coiled-coil homology domain . It is predominantly localized in the trans-Golgi network and endosomes, where it plays a pivotal role in the sorting and transport of proteins . The protein is also found in the plasma membrane, contributing to its diverse functional roles within the cell .

Functional Role

The primary function of Syntaxin-6 involves the regulation of intracellular vesicle trafficking. It interacts with other SNARE proteins, such as STX12, VAMP4, and VTI1A, to form a complex that facilitates the fusion of vesicles with target membranes . This interaction is essential for the proper delivery of cargo proteins to their designated locations within the cell.

Clinical Significance

Syntaxin-6 has been implicated in various pathological conditions, particularly in the context of cancer. Overexpression of STX6 has been observed in several types of human malignant tumors, including esophageal, colorectal, and renal cell carcinomas . Recent studies have highlighted its role in hepatocellular carcinoma (HCC), where it promotes tumor progression by enhancing the activation of the JAK-STAT signaling pathway . This makes Syntaxin-6 a potential therapeutic target for cancer treatment.

Recombinant Human Syntaxin-6

Recombinant human Syntaxin-6 is produced using conventional chromatography techniques and is typically expressed in E. coli . The recombinant protein is often tagged with a His-tag at the N-terminus to facilitate purification and detection. It is used extensively in research to study the protein’s function, interactions, and role in disease mechanisms .

Applications in Research

Recombinant human Syntaxin-6 is utilized in various experimental applications, including:

  • Protein-Protein Interaction Studies: To investigate the interactions between Syntaxin-6 and other SNARE proteins or regulatory factors.
  • Functional Assays: To assess the role of Syntaxin-6 in vesicle trafficking and membrane fusion events.
  • Cancer Research: To explore the involvement of Syntaxin-6 in tumorigenesis and its potential as a therapeutic target.

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.