STX11 Human

Syntaxin-11 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Vesicle Exocytosis in Immune Cells

STX11 mediates SNARE complex assembly with SNAP-23 and VAMP proteins to enable cytotoxic granule fusion with the plasma membrane in CTLs and NK cells . Defects in STX11 impair degranulation, leading to familial hemophagocytic lymphohistiocytosis type 4 (FHL-4) .

Key Findings in Immune Function

ProcessSTX11’s RoleOutcome of Deficiency
Cytotoxic Granule ExocytosisForms SNARE complexes (SNAP-23, VAMP) for granule fusion Impaired perforin/granzyme release → FHL-4
CD4 T Cell-B Cell InteractionsMobilizes CD40L, IL-2, IL-10; supports germinal center formation Reduced class-switched antibodies, low IgG levels
TLR4 TransportCooperates with SNAP-23 to traffic TLR4 to the plasma membrane in macrophages Dysregulated innate immune responses

Regulation of Autophagy and Fibrosis

STX11 overexpression inhibits pulmonary fibrosis by promoting autophagy in human lung fibroblasts (HLFs) and suppressing the PI3K/AKT/mTOR pathway . This involves interactions with SNAP25, which reciprocally upregulates STX11 expression .

Familial Hemophagocytic Lymphohistiocytosis Type 4 (FHL-4)

Key Features of FHL-4

FeatureDescription
Genetic CauseBiallelic STX11 mutations (e.g., p.Leu194ProfsX2)
Clinical PresentationHyperinflammation, cytopenia, hepatosplenomegaly, elevated IFN-γ
Mouse Model InsightsReduced IgG, impaired germinal centers, and hypogammaglobulinemia

Pulmonary Fibrosis

STX11 downregulation is observed in idiopathic pulmonary fibrosis (IPF) and bleomycin (BLM)-induced models. Overexpression of STX11 via adeno-associated virus (AAV) reduces fibrosis severity by:

  • Suppressing α-SMA, fibronectin, and collagen I expression .

  • Blocking TGF-β1-induced fibroblast activation .

Recombinant STX11 Protein

Recombinant STX11 is used to study:

  • Fibroblast Autophagy: Inhibits HLF activation by TGF-β1 .

  • SNARE Interactions: Forms complexes with SNAP25 to regulate membrane fusion .

Mouse Models

ModelFindings
Stx11−/− MiceImpaired degranulation, reduced IgG, increased IgM, and FHL-like symptoms
AAV-STX11 OverexpressionAlleviates BLM-induced fibrosis via decreased α-SMA and collagen

Protein Interactions

PartnerInteractionFunctional Impact
SNAP-23Forms SNARE complexes for granule fusion Enables cytotoxicity in immune cells
SNAP-25Reciprocal upregulation; inhibits PI3K/AKT/mTOR in fibroblasts Reduces fibrosis and promotes autophagy
TLR4Cooperates with SNAP-23 for TLR4 trafficking to plasma membrane Modulates innate immune responses

Pathway Inhibition

STX11 inhibits the PI3K/AKT/mTOR pathway, which is critical for fibroblast activation and proliferation . This inhibition is mediated through its interaction with SNAP25 .

Product Specs

Introduction
Syntaxin-11 (STX11) is a member of the t-SNARE family of proteins. It plays a crucial role in regulating protein trafficking between late endosomes and the trans-Golgi network. STX11 interacts with other SNARE proteins, including SNAP-23 and VAMP, to facilitate this transport process. Mutations in the STX11 gene have been linked to the development of familial hemophagocytic lymphohistiocytosis.
Description
This product consists of recombinant human STX11 protein produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 311 amino acids (specifically, amino acids 1-287) and has a molecular weight of 35.8 kDa. The STX11 protein is fused to a 24 amino acid His-tag at the N-terminus to facilitate purification, which is carried out using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The STX11 protein is supplied in a solution at a concentration of 1 mg/ml. The solution also contains 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For longer storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (such as 0.1% HSA or BSA). Avoid subjecting the product to multiple freeze-thaw cycles.
Purity
The purity of the STX11 protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Syntaxin-11, STX11, FHL4, HLH4, HPLH4.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMKDRLA ELLDLSKQYD QQFPDGDDEF DSPHEDIVFE TDHILESLYR DIRDIQDENQ LLVADVKRLG KQNARFLTSM RRLSSIKRDT NSIAKAIKAR GEVIHCKLRA MKELSEAAEA QHGPHSAVAR ISRAQYNALT LTFQRAMHDY NQAEMKQRDN CKIRIQRQLE IMGKEVSGDQ IEDMFEQGKW DVFSENLLAD VKGARAALNE IESRHRELLR LESRIRDVHE LFLQMAVLVE KQADTLNVIE LNVQKTVDYT GQAKAQVRKA VQYEEKNPCR TLCCFCCPCL K.

Q&A

What is STX11 and what is its primary function in human cells?

STX11 (Syntaxin 11) is a t-SNARE (target-membrane soluble N-ethylmaleimide-sensitive factor attachment protein receptor) protein involved in membrane fusion events. In human cells, STX11 serves as a critical mediator of lytic granule fusion, particularly in cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. The protein facilitates the docking and fusion of vesicles with target membranes, playing an essential role in immune cell cytotoxicity. STX11 functions by forming complexes with other SNARE proteins to create a fusion-competent complex that drives membrane merger. High-resolution imaging has revealed that STX11 clusters at the immunological synapse where it serves as a platform for arriving lytic granules . This molecular function explains why STX11 deficiency severely compromises immune cell cytotoxicity, leading to immune dysregulation.

What diseases are associated with STX11 mutations in humans?

The primary clinical manifestation of STX11 mutations is Familial Hemophagocytic Lymphohistiocytosis type 4 (FHL-4), a rare but potentially fatal autoimmune disorder. FHL-4 belongs to a group of primary immunodeficiency disorders characterized by hyperinflammation and immune dysregulation. Patients with STX11 mutations present with fever, splenomegaly, bicytopenia, hypertriglyceridemia, hypofibrinogenemia, hemophagocytosis, low/absent NK cell activity, hyperferritinemia, and elevated soluble interleukin-2 receptor levels . The disease is autosomal recessive, often emerging in children from consanguineous marriages with homozygous STX11 mutations. Various mutation types have been documented, including missense/nonsense mutations, minor deletions, and large fragment deletions . The phenotypic presentation can vary considerably, even among patients with identical STX11 genotypes, suggesting that additional genetic or environmental factors influence disease expression.

How does STX11 interact with other proteins in the SNARE complex?

STX11 forms functional complexes with several key proteins to facilitate membrane fusion events. One critical interaction is with Munc18-2 (also known as STXBP2), which regulates STX11 function and availability. Research indicates that both the N-terminus and Habc domain of STX11 are required for binding to Munc18-2, suggesting a mechanism similar to the dynamic binary binding observed between neuronal syntaxin-1 and Munc18-1 .

STX11 also interacts with SNAP proteins, including SNAP23 and SNAP25, to form functional SNARE complexes. Molecular dynamics simulations have revealed specific interaction points between STX11 and other proteins. For example, the Habc domain of STX11 forms stable interactions with the C-terminus of Orai1, a calcium channel component. These interactions include salt bridges between specific amino acid residues (Arg78_STX11 and Glu275_Orai1, Glu150_STX11 and Arg289_Orai1, Arg160_STX11 and Glu272_Orai1), hydrogen bonds (between Asn147_STX11 and Arg289_Orai1, Gln164_STX11 and Glu272_Orai1), and potential cation-pi interactions (between Tyr146_STX11 and Arg289-Orai1) . These molecular interactions highlight the sophisticated binding interface that enables STX11's function in membrane fusion events.

What is the structure of the STX11 gene and protein?

The STX11 gene in humans contains multiple exons that encode a protein of approximately 287 amino acids with a molecular weight of approximately 33 kDa. The protein contains several functional domains that are critical for its activity:

  • N-terminal region: Important for binding to regulatory proteins like Munc18-2

  • Habc domain: A three-helix bundle that mediates protein-protein interactions and regulates SNARE complex formation

  • SNARE motif: The core functional domain that participates in the formation of the SNARE complex

  • C-terminal region: Involved in membrane targeting and protein interactions

Unlike many other syntaxins, STX11 lacks a transmembrane domain and instead associates with membranes through post-translational lipid modifications and protein-protein interactions. This structural feature may contribute to its specialized function in immune cells. Mutations affecting different domains of STX11 have varying impacts on protein function. For example, the p.Q17X mutation causes premature termination of protein synthesis, resulting in the loss of 240 amino acids and severely compromised function .

What are the current methods for detecting STX11 mutations in research and clinical settings?

Detection of STX11 mutations requires a comprehensive genetic approach that typically follows this methodological workflow:

  • DNA Extraction: Genomic DNA is isolated from peripheral blood mononuclear cells (PBMCs) of patients.

  • PCR Amplification: The STX11 gene's exons, including 5' UTR, coding regions, and exon-intron boundaries, are amplified using specific primer sets. The reaction typically contains 2.5 mM MgCl₂, 200 nM dNTPs, 250 nM of both forward and reverse primers, and 0.5 unit of DNA polymerase. The amplification protocol generally consists of 35 cycles (96°C/30s, 55°C/30s, 72°C/30s) followed by a final extension at 72°C for 7 minutes .

  • PCR Product Processing: PCR products are treated with ExoSAP-IT to inactivate free primers and dNTPs, then purified using magnetic bead-based methods.

  • Sequencing: Bidirectional Sanger sequencing is performed to obtain reliable sequence data. Advanced clinical laboratories may use next-generation sequencing (NGS) panels that include STX11 along with other HLH-associated genes.

  • Sequence Analysis: Sample sequences are aligned to reference genomic sequences using programs like ClastalW to identify nucleotide changes. Variants are then classified according to ACMG guidelines.

For functional validation of identified variants, researchers employ additional techniques including Western blotting to assess protein expression, co-immunoprecipitation to evaluate protein-protein interactions, and cell-based assays to measure NK cell and CTL degranulation (measuring CD107a expression by flow cytometry) .

How can researchers effectively study STX11 function in primary immune cells?

Studying STX11 function in primary immune cells presents unique challenges due to their limited availability and short lifespan in culture. A comprehensive methodological approach includes:

  • Cell Isolation and Culture:

    • Isolate PBMCs from whole blood using density gradient centrifugation

    • Enrich NK cells or T cells using magnetic bead-based negative selection

    • Culture cells in appropriate media (RPMI-1640 with 10% FBS for T cells) supplemented with cytokines (IL-2 at 100 IU/ml for NK cells)

  • Gene Manipulation Strategies:

    • For transient knockdown: siRNA or shRNA delivery via nucleofection (Amaxa technology)

    • For stable knockdown: lentiviral vectors containing shRNA sequences targeting STX11

    • For overexpression: Transfection of expression plasmids containing wild-type or mutant STX11 constructs

    • For gene editing: CRISPR-Cas9 targeting of STX11 locus

  • Functional Assays:

    • Degranulation Assay: Measure surface expression of CD107a (LAMP-1) by flow cytometry following target cell stimulation or PMA/ionomycin treatment

    • Cytotoxicity Assays: Chromium release assay or flow cytometry-based killing assays using labeled target cells

    • Calcium Flux: Measure store-operated calcium entry (SOCE) using fluorescent calcium indicators

    • Vesicle Trafficking: Advanced microscopy techniques including total internal reflection fluorescence (TIRF) microscopy to visualize granule movement and fusion events

  • Protein Interaction Studies:

    • Co-immunoprecipitation to identify binding partners

    • Proximity ligation assays to visualize protein interactions in situ

    • FRET/FLIM to measure direct protein-protein interactions in living cells

These methodologies can be combined with patient-derived cells carrying STX11 mutations to establish genotype-phenotype correlations and validate experimental findings in a disease-relevant context .

What experimental approaches can reveal the molecular mechanism of STX11 in membrane fusion?

Understanding STX11's molecular mechanism in membrane fusion requires sophisticated experimental approaches:

  • High-Resolution Imaging Techniques:

    • Dual-color TIRF microscopy: This technique provides high temporal and spatial resolution of membrane-proximal events. By labeling STX11 and lytic granules with different fluorophores, researchers can visualize their dynamics at the immunological synapse. This approach has revealed that STX11 is first transported to the immunological synapse through recycling endosomes, where it forms clusters that serve as docking platforms for arriving lytic granules .

    • Super-resolution microscopy: Techniques like STORM or PALM can provide nanoscale resolution of STX11 clustering and SNARE complex formation.

    • Live-cell imaging: Using fluorescently tagged proteins to track real-time dynamics of STX11 and interacting partners.

  • Protein-Protein Interaction Analysis:

    • Pull-down assays: Using recombinant proteins to identify direct binding partners. For example, MBP-tagged Orai1 fragments can be used to pull down His-tagged STX11 to map interaction domains .

    • Surface plasmon resonance (SPR): To measure binding kinetics and affinity between STX11 and its partners.

    • Molecular dynamics simulations: Computational approaches to model protein interactions. This has been used to assess the stability of STX11 Habc domain interaction with Orai1 C-terminus, revealing specific salt bridges and hydrogen bonds .

  • Functional Reconstitution Systems:

    • Liposome fusion assays: In vitro reconstitution of membrane fusion using purified components to directly test STX11's fusion capability.

    • Cell-free granule fusion assays: Isolated granules and plasma membrane preparations to study the minimal machinery required for fusion.

  • Structure-Function Analysis:

    • Domain mapping: Creation of truncation mutants to identify functional regions, as demonstrated with STX11 R4A and L58P mutations that revealed the importance of both N-terminus and Habc domain for Munc18-2 binding .

    • Point mutations: Site-directed mutagenesis to test the importance of specific residues.

    • Chimeric proteins: Swapping domains between STX11 and other syntaxins to identify unique functional elements.

The combination of these approaches has helped establish STX11 as a bona fide t-SNARE that mediates the final fusion step of lytic granules at the immunological synapse .

How do STX11 mutations influence the development and progression of hemophagocytic lymphohistiocytosis?

STX11 mutations impact hemophagocytic lymphohistiocytosis (HLH) development through several mechanisms that can be categorized in a functional framework:

  • Defective Cytotoxicity Pathway:
    STX11 mutations directly impair the fusion of lytic granules with the plasma membrane in NK cells and CTLs. This results in failed target cell killing, which disrupts immune homeostasis. Without effective cytotoxicity, activated lymphocytes persist and continue to secrete pro-inflammatory cytokines, leading to uncontrolled inflammation. Quantitative assessment of this defect shows significantly reduced degranulation (ΔMFI: <1.4%, compared to normal ≥2.8%) as measured by CD107a expression .

  • Temporal Disease Progression:
    The correlation between mutation type and disease onset/progression shows substantial heterogeneity. In a study of Turkish families with STX11 mutations, onset age ranged from 1 month to 7 years, with variable disease severity and response to treatment. Notably, some patients with identical mutations (p.Q268X) presented at dramatically different ages (1 month versus 7 years), indicating that additional factors influence disease expression . This phenotypic variability suggests compensatory mechanisms may exist, as evidenced by a patient with a severe Stx11 protein defect (p.Q17X) who nonetheless had late disease onset (4.5 years) and long remission periods .

  • Calcium Signaling Dysfunction:
    Recent research has uncovered a previously unknown role for STX11 in regulating store-operated calcium entry (SOCE). STX11-deficient cells show impaired calcium influx, which further compromises immune cell function. In patient-derived cells with a frameshift mutation in STX11 (deletion of adenine at position 752), SOCE was significantly reduced compared to healthy controls, and this defect could be reversed by expression of wild-type STX11 .

  • Triggering Factors:
    While STX11 mutations create susceptibility, viral infections (particularly EBV) often trigger disease manifestation. Careful monitoring of EBV DNA in patient plasma can help predict and potentially prevent disease recurrence .

This multifaceted understanding of how STX11 mutations influence HLH has direct implications for diagnosis, treatment timing, and the development of targeted therapies that might bypass the defective cytotoxicity pathway.

What is the relationship between STX11 and calcium signaling in immune cells?

The relationship between STX11 and calcium signaling in immune cells represents a newly discovered mechanism with profound implications for immune function:

  • Role in CRAC Channel Regulation:
    STX11 directly interacts with the Orai1 protein, which forms the pore of the calcium release-activated calcium (CRAC) channel. Specific molecular dynamics simulations have identified key interaction points between STX11's Habc domain and Orai1's C-terminus. This interaction is characterized by stable salt bridges, hydrogen bonds, and potential cation-pi interactions between specific residues .

  • Mechanistic Impact on Store-Operated Calcium Entry (SOCE):
    When STX11 is depleted, several critical calcium signaling defects emerge:

    • Decreased peak cytosolic calcium levels following store depletion

    • Reduced CRAC current densities

    • Impaired functional entrapment of Orai1 by STIM1

    • Defective channel gating even when Orai1 and STIM1 are artificially co-clustered

    These findings indicate that STX11 is not merely required for physical association of STIM1 and Orai1 but plays a fundamental role in channel priming and activation .

  • Experimental Evidence from Patient Cells:
    In T cells from a familial hemophagocytic lymphohistiocytosis type 4 (FHLH4) patient with a STX11 mutation (deletion of adenine at position 752), SOCE was significantly reduced compared to healthy controls. This defect was reversed by reconstitution with wild-type STX11, conclusively demonstrating STX11's direct role in calcium signaling .

  • Distinction from Other SNARE Functions:
    This calcium regulation function appears distinct from STX11's canonical role in membrane fusion, as other SNARE proteins (SNAP23, SNAP25, and SNAP29) do not show the same effect on SOCE when depleted. This suggests a specialized function for STX11 in immune cell calcium homeostasis beyond its role in lytic granule exocytosis .

This relationship between STX11 and calcium signaling has significant implications for understanding immune cell activation, as calcium influx is a critical second messenger for numerous signaling pathways controlling cytokine production, proliferation, and effector functions.

What are the current therapeutic approaches for patients with STX11 mutations, and how can research advance treatment options?

Current therapeutic approaches for patients with STX11 mutations focus on managing the resulting hemophagocytic lymphohistiocytosis (HLH), with a progressive strategy from symptom control to definitive treatment:

  • Initial Management Using HLH-2004 Protocol:

    • Immunosuppression: Dexamethasone and etoposide to control hyperinflammation

    • Cyclosporine: Used for maintenance therapy to prevent disease reactivation

    • Supportive Care: Management of cytopenias, coagulopathy, and organ dysfunction

    Patient data shows this approach can achieve remission, as demonstrated in the case of a patient with STX11 p.Q17X mutation who maintained disease control for over 6 years with cyclosporine maintenance therapy .

  • Definitive Therapy:

    • Hematopoietic Stem Cell Transplantation (HSCT): Currently the only curative option for patients with STX11 mutations

    • HLA Matching: Essential for successful transplantation outcomes

    • Donor Selection: Related or unrelated donors with appropriate HLA matching

  • Research Directions to Advance Treatment Options:

    • Gene Therapy Approaches: Development of viral vectors to deliver functional STX11 to patient cells

    • CRISPR-Cas9 Gene Editing: Correction of specific STX11 mutations in hematopoietic stem cells

    • Small Molecule Modulators: Identification of compounds that could bypass STX11 deficiency by:

      • Directly activating downstream membrane fusion machinery

      • Enhancing calcium signaling to overcome associated SOCE defects

      • Promoting alternative degranulation pathways in NK cells and CTLs

  • Precision Medicine Strategies:

    • Mutation-Specific Approaches: Tailoring therapy based on the specific STX11 mutation and its functional consequences

    • Compensatory Mechanism Enhancement: Identification and therapeutic augmentation of natural compensatory pathways, as suggested by the variability in disease onset and progression even among patients with similar mutations

    • Biomarker Development: Identification of early markers for disease activation to guide preemptive therapy

  • Novel Therapeutic Targets Based on Recent Discoveries:

    • SOCE Modulation: Development of therapies targeting the STX11-Orai1 interaction to restore calcium signaling

    • Alternative SNARE Complex Formation: Promoting formation of functional SNARE complexes despite STX11 deficiency

These research directions offer promising avenues for developing more targeted, effective, and potentially less toxic therapeutic options beyond the current standard of care for patients with STX11 mutations.

How does STX11 function beyond the immune system, and what are its potential roles in other tissues?

Recent discoveries have expanded our understanding of STX11 function beyond the immune system, revealing diverse roles in multiple tissues:

  • Pulmonary Fibrosis Regulation:
    A groundbreaking study has revealed that STX11 plays a protective role against pulmonary fibrosis. Overexpression of STX11 alleviates fibrotic changes through interaction with SNAP25. This STX11-SNAP25 complex inhibits lung fibrosis through two primary mechanisms:

    • Promotion of fibroblast autophagy

    • Suppression of fibroblast activation

    This discovery suggests STX11 as a potential therapeutic target for fibrotic lung diseases, significantly expanding its known physiological importance beyond immune cells .

  • Vesicle Trafficking in Non-Immune Cells:
    STX11's role as a t-SNARE protein suggests it may participate in membrane fusion events in various cell types. Research indicates potential functions in:

    • Regulated exocytosis in secretory epithelia

    • Autophagosome-lysosome fusion

    • Vesicular transport within the Golgi apparatus

    These functions remain less well-characterized than its role in immune cells but represent important areas for future research.

  • Metabolic Regulation:
    Emerging evidence suggests STX11 may participate in glucose metabolism through:

    • Regulation of GLUT4 vesicle trafficking in response to insulin

    • Modulation of mitochondrial dynamics and function

    • Potential crosstalk with cellular energy sensing pathways

    These metabolic functions may connect STX11 to conditions beyond primary immunodeficiency.

  • Developmental Processes:
    The spatiotemporal expression pattern of STX11 during development suggests roles in:

    • Tissue morphogenesis

    • Cell differentiation pathways

    • Establishment of tissue architecture

    Further research is needed to define these developmental functions and their potential contribution to congenital disorders.

These diverse functions highlight STX11 as a multifunctional protein with significance extending far beyond its established role in immune cell cytotoxicity. Understanding these pleiotropic effects will provide a more comprehensive picture of how STX11 mutations lead to disease and may uncover novel therapeutic approaches for multiple conditions.

What are the most effective experimental models for studying STX11 function and related diseases?

Selecting appropriate experimental models for studying STX11 function requires careful consideration of the specific research questions and the translational relevance. A comprehensive approach utilizes multiple complementary models:

  • Cellular Models:

    Cell TypeAdvantagesLimitationsBest Applications
    Primary human NK cellsPhysiologically relevant, disease-specific mutations can be studiedLimited availability, short lifespan, difficult to genetically manipulateValidation of findings, patient-specific studies
    Primary T lymphocytesCan be expanded in culture, relevant for cytotoxicity studiesVariable donor characteristics, activation state affects resultsFunctional assays, calcium signaling studies
    NK cell lines (NK-92, YTS)Homogeneous population, amenable to genetic manipulationMay not recapitulate all aspects of primary NK cell biologyMechanistic studies, high-throughput screening
    HEK293 cellsEasily transfected, suitable for recombinant protein expressionNot immune cells, lack specialized secretory lysosomesProtein-protein interaction studies, structure-function analysis
    Jurkat T cellsT cell lineage, easy to culture and manipulateTransformed cells with altered signaling pathwaysCalcium signaling, CRAC channel function
  • Animal Models:

    ModelAdvantagesLimitationsBest Applications
    STX11 knockout miceComplete absence of protein, systemic effects can be studiedSpecies differences in immune functionIn vivo pathophysiology, organ-specific manifestations
    Conditional STX11 knockoutCell type-specific deletion, temporal controlComplex breeding schemes, potential compensationDissecting cell-autonomous effects
    Humanized mouse modelsHuman immune system in vivoIncomplete human immune development, expenseTesting human-specific therapeutics
    CRISPR-engineered models with specific patient mutationsPrecise recapitulation of human mutationsTime-consuming generation, potential off-target effectsGenotype-phenotype correlations
    Zebrafish modelsRapid development, optical transparency for imagingEvolutionary distance from humansHigh-throughput screening, developmental studies
  • Advanced Complex Models:

    • Organoids: 3D culture systems that better recapitulate tissue architecture and cellular interactions

    • Microfluidic systems: Allow precise control of cell-cell interactions and imaging of immune synapse formation

    • Patient-derived iPSCs: Can be differentiated into relevant cell types while maintaining patient-specific genetic background

    • Ex vivo tissue cultures: Maintain the complex multicellular environment of organs like lymph nodes or spleen

  • Computational Models:

    • Molecular dynamics simulations: For studying STX11 protein interactions at atomic resolution

    • Systems biology approaches: To understand STX11 in the context of broader signaling networks

    • Machine learning algorithms: To identify patterns in patient data and predict disease course

The most effective research strategy often involves a multi-model approach, starting with simpler systems to establish mechanisms and moving to more complex models for validation and translational relevance. For instance, a mechanistic finding in HEK293 cells about STX11-Orai1 interaction can be validated in primary T cells and then assessed for physiological relevance in conditional knockout mice .

What are the unresolved questions regarding STX11 regulation and function that warrant further investigation?

Despite significant advances in understanding STX11, several critical questions remain unresolved and represent important areas for future research:

  • Regulatory Mechanisms of STX11 Expression and Activity:

    • What transcription factors and epigenetic mechanisms control STX11 expression in different cell types?

    • How is STX11 activity regulated post-translationally (phosphorylation, ubiquitination, etc.)?

    • What are the mechanisms of STX11 targeting to specific membrane compartments without a transmembrane domain?

    • How is STX11 function modulated during different phases of immune cell activation?

  • Molecular Structure and Dynamics:

    • What is the complete three-dimensional structure of STX11 alone and in complex with binding partners?

    • How does STX11 conformation change during SNARE complex assembly and membrane fusion?

    • What are the structural determinants that make STX11 unique among syntaxin family members?

    • How do disease-causing mutations specifically alter STX11 structure and dynamics?

  • Non-canonical Functions:

    • What is the detailed molecular mechanism by which STX11 regulates calcium signaling via Orai1?

    • Does STX11 play roles in autophagy, mitophagy, or other cellular quality control mechanisms?

    • How does STX11 contribute to cell motility, polarization, and migration?

    • What are STX11's functions in non-immune tissues during development and homeostasis?

  • Disease Heterogeneity and Modifiers:

    • What accounts for the variable disease onset and severity among patients with identical STX11 mutations?

    • Are there genetic modifiers that influence the phenotypic expression of STX11 mutations?

    • How do environmental factors, particularly viral infections, interact with STX11 deficiency?

    • What compensatory mechanisms exist in patients with milder disease courses despite significant STX11 dysfunction?

  • Therapeutic Development:

    • Can small molecules enhance residual STX11 function in patients with hypomorphic mutations?

    • Is it possible to bypass STX11 deficiency by targeting downstream effectors?

    • What are the long-term outcomes and optimal conditioning regimens for HSCT in STX11-deficient patients?

    • Could STX11 gene therapy provide a viable alternative to HSCT?

  • Emerging Connections:

    • What is the relationship between STX11 and metabolism, particularly in the context of inflammatory diseases?

    • How does STX11 interact with the cytoskeleton during immune synapse formation?

    • Does STX11 play a role in trained immunity or memory-like responses in innate immune cells?

    • What is the contribution of STX11 to age-related immune dysfunction?

Addressing these questions will require innovative approaches combining structural biology, advanced imaging, systems biology, and integrative analysis of patient data. The answers promise to not only enhance our understanding of STX11 biology but also improve diagnosis and treatment of STX11-related disorders.

How can systems biology and multi-omics approaches advance our understanding of STX11 function in health and disease?

Systems biology and multi-omics approaches offer powerful frameworks for comprehensively understanding STX11 function within complex biological networks:

  • Network Analysis and Pathway Integration:
    Systems biology can reveal how STX11 functions within broader cellular networks by:

    • Mapping protein-protein interaction networks centered on STX11

    • Identifying feedback loops and regulatory circuits involving STX11

    • Modeling dynamic changes in these networks during immune cell activation

    • Predicting system-wide effects of STX11 mutations through computational simulation

    These approaches can uncover unexpected connections between STX11 and other cellular processes, explaining the pleiotropic effects of STX11 dysfunction.

  • Multi-omics Data Integration Strategies:
    Combining multiple omics datasets can provide unprecedented insight into STX11 biology:

    Omics ApproachKey Information ProvidedApplication to STX11 Research
    GenomicsMutation identification, population variationCataloging disease-causing variants, identifying genetic modifiers
    TranscriptomicsExpression patterns, co-regulated genesDefining STX11 expression networks across tissues and conditions
    ProteomicsProtein abundance, post-translational modificationsMapping STX11 interactome, identifying regulatory modifications
    PhosphoproteomicsSignaling pathway activationUnderstanding how STX11 is regulated by kinase networks
    MetabolomicsMetabolic pathway alterationsConnecting STX11 to cellular metabolism and energetics
    LipidomicsMembrane composition changesAnalyzing impact on membrane fusion and organization
    Single-cell omicsCellular heterogeneityIdentifying cell-specific roles and responses to STX11 deficiency

    Integration of these datasets using machine learning approaches can identify patterns and correlations not evident from any single dataset.

  • Clinical Translation of Systems Approaches:
    Systems biology can transform patient care by:

    • Developing predictive biomarkers for disease onset in carriers of STX11 mutations

    • Identifying patient-specific therapeutic targets based on personalized network analysis

    • Stratifying patients for clinical trials based on molecular signatures

    • Monitoring disease progression through dynamic changes in multi-omics profiles

  • Emerging Methodological Advances:
    New technologies that will accelerate systems-level understanding include:

    • Spatial transcriptomics/proteomics to map STX11 function in tissue context

    • Time-resolved omics to capture dynamic processes during immune cell activation

    • Single-molecule techniques to observe individual STX11 molecules during membrane fusion

    • Artificial intelligence approaches to integrate heterogeneous data types and predict emergent properties

These systems biology and multi-omics approaches promise to transform our understanding of STX11 from a single protein to a key node in complex cellular networks, potentially revealing novel therapeutic strategies for STX11-related disorders that target not just the protein itself but the broader dysregulated system.

Product Science Overview

Structure and Function

Syntaxin-11 is a membrane protein that is involved in the fusion of vesicles with target membranes. It forms part of the SNARE complex, which is essential for the docking and fusion of vesicles . The SNARE complex typically consists of syntaxins, SNAP-25 (synaptosome-associated protein of 25 kDa), and VAMPs (vesicle-associated membrane proteins). In the case of syntaxin-11, it has been shown to form complexes with VAMP-8 and SNAP-23 .

Role in Immune Function

Syntaxin-11 is critical for the proper functioning of immune cells. It is involved in the degranulation process of NK cells and CTLs, which is essential for the release of cytotoxic granules that kill target cells . Deficiencies or mutations in the STX11 gene can lead to impaired immune responses and are associated with a rare immune disorder known as familial hemophagocytic lymphohistiocytosis type 4 (FHL4) . This disorder is characterized by uncontrolled activation of immune cells, leading to severe inflammation and tissue damage.

Recombinant Human Syntaxin-11

Recombinant human syntaxin-11 is a synthetically produced version of the protein, often used in research to study its function and role in various cellular processes. It is typically expressed in E. coli and purified for use in experiments . The recombinant protein is often tagged with a His-tag at the N-terminus to facilitate purification and detection .

Research and Clinical Implications

Research on syntaxin-11 has provided valuable insights into its role in immune cell function and its involvement in immune disorders. Studies using syntaxin-11-deficient murine models have shown that the absence of this protein leads to defects in the degranulation process of immune cells, resulting in impaired cytotoxic activity . These findings have important implications for understanding the mechanisms underlying immune disorders and developing potential therapeutic strategies.

In conclusion, syntaxin-11 is a vital protein involved in the regulation of vesicle fusion and immune cell function. Recombinant human syntaxin-11 serves as an important tool in research, helping scientists to unravel the complexities of immune responses and develop new treatments for immune-related 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.