Recombinant Mouse Sorting nexin-8 (Snx8)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and serves as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is crucial for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The specific tag type is determined during production. If a particular tag type is required, please inform us, and we will prioritize its development.
Synonyms
Snx8; Sorting nexin-8
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-459
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Snx8
Target Protein Sequence
MTGRAMDPLP SPAVAAAAEA EADEEADPPA TGPRTSQVTE WRALDPGRMQ MPQGNPLLLS YTLQELLAKD TVQVELIPEK KGLFLKHVEY EVSSQRFKSS VYRRYNDFVV FHEVLLHKFP YRMVPALPPK RVLGADREFI EGRRRALKRF INLVARHPPF SEDVLLKLFL SFSGSDVQHK LKEAAQCVGD EFMNCKLAAR AKDFLPADIQ TQFAMSRELI RNVYNSFYKL RDRAERIASR AIDNAADLLI FGKELSALGS DTTPLPSWAA LHLSTWGSLK QALKGLSVEF ALLADRAAQQ GKKEENDVVE KLNLFLDLLQ SYKDLCERHE KGVLHKHQRA LHKYGLMKRQ MMSAAHGREP ESVEQLESRI VEQENVIQTM ELRNYFSLYC LHQETQLVHV YLPLTSHILG AFVNSQIQGH KEMSKVWNDL KPKLSCLFAG PHSVLTPPRS PQEDGVCPH
Uniprot No.

Target Background

Function
Potentially involved in multiple stages of intracellular trafficking. May play a role in intracellular protein transport from early endosomes to the trans-Golgi network.
Gene References Into Functions
  1. SNX8 mediates IFNγ-triggered non-canonical signaling and host defense against Listeria monocytogenes. PMID: 29180417
Database Links
Protein Families
Sorting nexin family
Subcellular Location
Early endosome membrane; Peripheral membrane protein; Cytoplasmic side.

Q&A

What is Sorting Nexin-8 (SNX8) and what are its primary functions in cellular processes?

Sorting nexin-8 (SNX8) is a member of the sorting nexin protein family characterized by the presence of a Phox homology (PX) domain that enables preferential association with endosomal membranes. SNX8 has multiple identified cellular functions, primarily in lysosome reformation and intracellular trafficking pathways. Recent research has demonstrated that SNX8 promotes lysosome tubulation, a critical process required for lysosome reformation when the functional lysosome pool is reduced . During this process, SNX8 helps recycle lysosomal lipids and proteins to restore lysosomal functions. Additionally, SNX8 participates in endosome-to-Golgi transport processes and colocalizes with early endosome antigen 1 (EEA1) and retromer components, confirming its endosomal localization . Beyond membrane trafficking, SNX8 plays significant roles in innate immune responses to RNA viruses and IFNγ-triggered signaling pathways .

How does SNX8 differ from other sorting nexin family members?

While all sorting nexins share the common PX domain, SNX8 possesses distinctive functional properties that differentiate it from other family members. Unlike many sorting nexins, SNX8 shows exceptional colocalization with LAMP1, a well-established lysosomal marker, indicating its specialized role in lysosomal functions . Experimental evidence demonstrates that when various sorting nexins are overexpressed, only SNX2 and SNX8 significantly colocalize with LAMP1, with SNX8 showing the highest degree of colocalization . Unlike some sorting nexins that form obligate heterodimers to function properly, SNX8 appears capable of independent function. Additionally, SNX8 plays unique roles in immune signaling that haven't been documented for other sorting nexin family members, such as its specific involvement in IFNγ-triggered noncanonical signaling pathways through interactions with JAK1 and IKKβ . These distinctions highlight SNX8's multifunctional nature compared to other sorting nexins.

What experimental approaches are commonly used to study SNX8 localization and interaction with other proteins?

Several established experimental techniques have proven effective for studying SNX8 localization and protein interactions. For subcellular localization studies, immunofluorescence microscopy using antibodies against endogenous SNX8 and organelle markers (particularly LAMP1 for lysosomes and EEA1 for early endosomes) is the primary approach . For dynamic localization studies, live cell imaging with fluorescently-tagged SNX8 has been employed to visualize its association with tubular lysosomal structures, especially under conditions like prolonged starvation that trigger lysosome reformation . Protein-protein interactions are typically investigated through co-immunoprecipitation assays, which have successfully identified SNX8's associations with JAK1, IKKβ, and VISA proteins . Additionally, biochemical fractionation methods can determine SNX8's membrane association properties, while yeast two-hybrid screening and mass spectrometry-based proteomics approaches have been used to identify novel SNX8 interaction partners.

What phenotypes are observed in SNX8-deficient cells and animal models?

SNX8 deficiency manifests in distinct cellular and organism-level phenotypes that reflect its diverse functions. At the cellular level, loss of SNX8 leads to phenotypes characteristic of Lysosomal Storage Disorders (LSDs), including enlarged lysosomes and defective lysosomal storage in human cells . These phenotypes suggest impaired lysosome reformation and function. In the context of immune responses, SNX8-deficient cells show impaired innate immune responses to RNA viruses, with reduced induction of downstream effector genes . At the organism level, Snx8−/− mice demonstrate increased susceptibility to lethal infection with RNA viruses compared to wild-type mice, highlighting SNX8's importance in antiviral immunity . Similarly, SNX8-deficient mice infected with Listeria monocytogenes exhibit lower serum cytokine levels and higher bacterial loads in the liver and spleen, resulting in increased mortality . These findings collectively demonstrate SNX8's critical roles in maintaining normal lysosomal function and mounting effective immune responses against pathogens.

How does SNX8 contribute to lysosome tubulation and reformation, and what experimental models best capture this function?

SNX8 facilitates lysosome tubulation through its membrane-binding and membrane-deforming capacities as a BAR domain-containing protein. During lysosome reformation, SNX8 localizes to tubular lysosomal structures and promotes the extension of these tubules, which eventually pinch off to form new functional lysosomes . This process is particularly active during prolonged starvation conditions when lysosome reformation is triggered. Live imaging analysis has revealed that both SNX2 and SNX8 localize onto tubular lysosomal structures under these conditions .

To effectively study this process, several experimental models have proven valuable:

Experimental ModelApplicationAdvantagesLimitations
Prolonged starvation in cultured cellsTriggers natural lysosome reformationPhysiologically relevant, easy to implementComplex cellular response beyond lysosomal effects
LAMP1-RFP transfected cells with GFP-SNX8Live visualization of lysosome tubulationReal-time dynamics, colocalization assessmentOverexpression artifacts possible
Hexb−/− mice (Sandhoff disease model)In vivo model of LSD for rescue experimentsAuthentic disease context, behavioral assessments possibleComplex pathophysiology, multiple cell types
AAV-SNX8 delivery to brainTherapeutic intervention assessmentTests translational potential, region-specific effectsDelivery efficiency variability, immune response to vector

The most effective approach combines these models, using cell culture systems to elucidate molecular mechanisms and mouse models to validate physiological relevance and therapeutic potential .

What is the mechanism by which SNX8 regulates the innate immune response to RNA viruses?

SNX8 regulates innate immune responses to RNA viruses through a specific molecular mechanism involving the mitochondrial antiviral signaling protein (VISA, also known as MAVS). Research has demonstrated that SNX8 physically associates with VISA and regulates its aggregation, which is an essential step for VISA activation and downstream signal transduction . The aggregation of VISA serves as a signaling platform that triggers the activation of transcription factors responsible for inducing antiviral genes.

Mechanistically, SNX8 functions as a positive regulator in this pathway, as gene knockout studies have confirmed that SNX8 deficiency significantly impairs the RNA virus-triggered induction of downstream effector genes . This regulation occurs at a post-receptor level in the signaling cascade. The biological significance of this regulation is substantial, as demonstrated by the increased susceptibility of Snx8−/− mice to lethal infection with RNA viruses compared to wild-type mice .

This mechanism represents a previously uncharacterized role for sorting nexins in antiviral immunity, expanding our understanding of how cellular trafficking proteins can have moonlighting functions in immune signaling pathways. The precise molecular events by which SNX8 promotes VISA aggregation require further investigation, but they likely involve SNX8's membrane-binding properties and potential scaffolding functions.

How does phosphorylation regulate SNX8 function in IFNγ-triggered signaling pathways?

Phosphorylation plays a critical regulatory role in modulating SNX8 function within IFNγ-triggered signaling pathways. Research has established that upon IFNγ stimulation, Janus kinase 1 (JAK1) phosphorylates SNX8 specifically at tyrosine residues Tyr95 and Tyr126 . This phosphorylation event is mechanistically significant as it promotes the recruitment of IKKβ (inhibitor of nuclear factor kappa-B kinase subunit beta) to the JAK1 complex.

The phosphorylation-dependent recruitment process follows a specific sequence:

  • IFNγ stimulation induces activation of JAK1/2

  • Activated JAK1/2 recruits SNX8 and phosphorylates it at Tyr95 and Tyr126

  • Phosphorylated SNX8 acts as a scaffold protein to recruit IKKβ to the JAK1 complex

  • This recruitment facilitates IKKβ dimerization/oligomerization and autophosphorylation at Ser177

  • Activated IKKβ selectively induces a subset of downstream genes important for host defense

Mutation of these phosphorylation sites (Tyr95 and Tyr126) to phenylalanine abolishes SNX8's ability to recruit IKKβ to the JAK1 complex, demonstrating the essential nature of these phosphorylation events for SNX8's signaling function . This phosphorylation-dependent mechanism highlights how post-translational modifications can confer specialized signaling functions to proteins primarily known for membrane trafficking roles, representing an elegant example of functional versatility through regulated protein modification.

What are the methodological considerations for generating and validating recombinant mouse SNX8 for experimental use?

Generating high-quality recombinant mouse SNX8 for experimental applications requires careful consideration of several methodological aspects:

Expression System Selection:
The choice of expression system significantly impacts protein quality and functionality. While bacterial systems (E. coli) offer high yield and cost-effectiveness, they lack post-translational modifications that may be important for SNX8 function. Mammalian expression systems (HEK293, CHO cells) provide proper folding and modifications but at lower yields. Insect cell systems (Sf9, Hi5) represent a middle ground, offering reasonable yields with eukaryotic post-translational processing.

Purification Strategy:
A multi-step purification approach is typically required:

  • Affinity chromatography using His-tag, GST-tag, or FLAG-tag as the initial capture step

  • Ion exchange chromatography for charge-based separation of contaminants

  • Size exclusion chromatography as a polishing step to ensure homogeneity

Quality Control Assessments:
Validation of recombinant SNX8 should include:

  • SDS-PAGE and Western blotting to confirm identity and purity

  • Mass spectrometry to verify sequence integrity and identify potential post-translational modifications

  • Circular dichroism to assess secondary structure content

  • Dynamic light scattering to evaluate homogeneity and aggregation status

  • Functional assays to confirm biological activity:

    • Membrane binding assays using liposomes containing phosphoinositides

    • In vitro tubulation assays to assess membrane deformation capacity

    • Protein interaction assays with known binding partners (JAK1, IKKβ)

Storage Considerations:
Optimizing storage conditions is crucial for maintaining activity:

  • Buffer composition (typically 20-50 mM Tris or phosphate, 100-150 mM NaCl, pH 7.4-8.0)

  • Addition of stabilizing agents (5-10% glycerol, 1-2 mM DTT)

  • Flash-freezing in liquid nitrogen and storage at -80°C in small aliquots to avoid freeze-thaw cycles

Thorough validation through these approaches ensures that experimental outcomes reflect authentic SNX8 biology rather than artifacts of improperly folded or modified recombinant protein.

How can recombinant SNX8 be utilized to investigate lysosomal storage disorders (LSDs)?

Recombinant SNX8 serves as a valuable tool for investigating the molecular mechanisms and potential therapeutic strategies for lysosomal storage disorders (LSDs). LSDs are characterized by enlarged lysosomes and defective lysosomal storage due to mutations in lysosome-related genes . Recent research has established that loss of SNX8 leads to phenotypes characteristic of LSDs in human cells, while SNX8 overexpression can rescue these features .

Several research applications of recombinant SNX8 in LSD investigations include:

  • Rescue experiments: Introduction of recombinant SNX8 into cells from LSD patients or LSD model organisms can help evaluate its therapeutic potential. For example, SNX8 overexpression successfully rescued the storage of GM2 ganglioside and the loss of NeuN+ neurons in Hexb−/− mice (a model of Sandhoff disease) .

  • Mechanism dissection: Using structure-function variants of recombinant SNX8 (with domain deletions or point mutations) helps identify which domains and residues are essential for its therapeutic effect in LSDs.

  • Interactome mapping: Recombinant SNX8 can be used in pull-down assays to identify binding partners in normal versus LSD cells, potentially revealing how LSD pathology alters SNX8's interaction network.

  • Drug screening platforms: Systems using fluorescently-tagged recombinant SNX8 can screen for small molecules that enhance SNX8's lysosome reformation activity or its binding to critical partners.

These applications collectively provide insights into both the basic biology of lysosomal homeostasis and potential therapeutic interventions for LSDs based on enhancing lysosome reformation pathways.

What experimental approaches can determine if SNX8 interacts with retromer components for endosomal sorting?

Determining SNX8's interactions with retromer components requires a multi-faceted experimental approach that combines imaging, biochemical, and functional assays. The retromer complex, consisting of a VPS26-VPS29-VPS35 heterotrimer and associated sorting nexins, plays crucial roles in endosomal sorting . Several experimental strategies can effectively assess SNX8's potential involvement with this complex:

  • Colocalization analysis: Confocal microscopy using fluorescently-tagged or antibody-detected SNX8 and retromer components (VPS26, VPS29, VPS35) can determine their spatial overlap in cells. Pearson's correlation coefficient and Manders' overlap coefficient provide quantitative measures of colocalization. Previous research has already shown that SNX8 colocalizes with retromer components, suggesting an endosomal localization .

  • Co-immunoprecipitation (Co-IP): This biochemical approach can identify physical interactions between SNX8 and retromer components. Both forward (immunoprecipitating SNX8 and blotting for retromer proteins) and reverse (immunoprecipitating retromer components and blotting for SNX8) approaches should be performed.

  • Proximity labeling methods: BioID or APEX2 fused to SNX8 can biotinylate proteins in close proximity in living cells, potentially identifying transient or weak interactions with retromer components that might be missed by Co-IP.

  • Yeast two-hybrid screening: This approach can test direct binary interactions between SNX8 and individual retromer components.

  • Functional cargo trafficking assays: Tracking the trafficking of known retromer-dependent cargoes (e.g., CI-MPR, sortilin) in cells with SNX8 depletion or overexpression can reveal functional relationships.

  • Structure-function analysis: Creating SNX8 variants with mutations in regions predicted to interact with retromer components can identify specific residues required for these interactions.

By combining these complementary approaches, researchers can establish not only whether SNX8 interacts with retromer components but also the nature, dynamics, and functional significance of these interactions in endosomal sorting processes.

How does SNX8 modulate autophagy and what methodologies are most effective for studying this relationship?

While direct evidence for SNX8's role in autophagy is emerging, its established functions in lysosome reformation and endosomal trafficking suggest potential involvement in autophagy regulation. Lysosomes are essential for the final degradative step of autophagy, and impaired lysosome function or reformation could significantly impact autophagic flux. To investigate this relationship, researchers can employ several specialized methodologies:

  • Autophagic flux assays: The most definitive approach to assess how SNX8 affects autophagy involves measuring autophagic flux using:

    • LC3-II turnover assays with and without lysosomal inhibitors (bafilomycin A1, chloroquine)

    • Tandem fluorescent-tagged LC3 (mRFP-GFP-LC3) to distinguish autophagosomes from autolysosomes

    • Long-lived protein degradation assays to measure autophagy-dependent proteolysis

  • Starvation response analysis: Since SNX8 localizes to tubular lysosomal structures under prolonged starvation , examining how SNX8 depletion or overexpression affects starvation-induced autophagy provides valuable insights. Comparing autophagic markers between fed and starved conditions in control versus SNX8-manipulated cells can reveal condition-specific effects.

  • Correlative light and electron microscopy (CLEM): This advanced technique enables visualization of SNX8's localization in relation to autophagic structures at ultrastructural resolution.

  • Selective autophagy monitoring: Assessing specific forms of selective autophagy (mitophagy, ER-phagy, xenophagy) in the context of SNX8 manipulation can reveal pathway-specific effects.

  • Genetic interaction studies: Creating double knockout/knockdown models combining SNX8 depletion with deficiencies in core autophagy genes (ATG5, ATG7, BECN1) can reveal epistatic relationships and position SNX8 in the autophagy pathway hierarchy.

  • In vivo autophagy assessment: Examining autophagy markers in tissues from SNX8-deficient mice, particularly under stressful conditions that induce autophagy, can validate cell culture findings in physiologically relevant contexts.

By systematically employing these methodologies, researchers can establish both if and how SNX8 modulates autophagy, potentially uncovering new therapeutic targets for disorders with dysregulated autophagy.

What considerations are important when assessing SNX8's role in different mouse tissues and disease models?

Evaluating SNX8's role across different mouse tissues and disease models requires careful consideration of several critical factors to ensure valid and interpretable results:

  • Tissue-specific expression patterns:
    SNX8 expression varies significantly across tissues, which necessitates baseline characterization of its endogenous expression levels. Quantitative PCR, Western blotting, and immunohistochemistry should be performed across major organ systems before undertaking tissue-specific functional studies. This baseline data informs expectations about where SNX8 manipulation would likely show the strongest phenotypes.

  • Disease model selection:
    The choice of disease model should align with SNX8's established functions. Based on existing research, several models are particularly relevant:

    • Lysosomal storage disorders: Hexb−/− mice (Sandhoff disease model) have already demonstrated SNX8's therapeutic potential

    • Viral infection models: RNA virus challenges in SNX8-deficient mice reveal its role in antiviral immunity

    • Bacterial infection models: Listeria monocytogenes infection demonstrates SNX8's importance in antibacterial defense

    • Neurodegenerative conditions: Given SNX8's role in lysosomal function, models of Alzheimer's, Parkinson's, or other neurodegenerative diseases may reveal additional functions

  • Genetic background considerations:
    The genetic background of mouse models can significantly influence phenotypes. Backcrossing SNX8 mutant mice to multiple pure backgrounds (C57BL/6, BALB/c, etc.) may reveal background-dependent effects and ensure reproducibility.

  • Compensatory mechanisms:
    Potential redundancy with other sorting nexins, particularly SNX2 which also localizes to lysosomes , must be addressed. Studies involving SNX8 should consider the expression of related sorting nexins that might compensate for its loss. Double knockout approaches may be necessary, similar to studies showing redundancy between SNX1 and SNX2 .

  • Temporal considerations:
    Developmental timing of SNX8 manipulation is crucial. Constitutive knockouts may trigger compensatory mechanisms, while inducible systems allow temporal control. For therapeutic applications, determining the optimal intervention window is essential, as demonstrated in AAV-SNX8 delivery to neonatal Hexb−/− mice .

  • Assessment methods:
    Multi-parametric assessment combining molecular, cellular, physiological, and behavioral measures provides the most comprehensive understanding of SNX8's role in disease models. This should include:

    • Molecular markers (lysosomal enzyme activities, cytokine profiles)

    • Cellular pathology (storage material accumulation, ultrastructural changes)

    • Physiological parameters (organ function tests)

    • Behavioral assessments (particularly for neurological conditions)

By systematically addressing these considerations, researchers can develop a nuanced understanding of SNX8's tissue-specific functions and therapeutic potential across multiple disease contexts.

What are the optimal conditions for expressing and purifying functional recombinant mouse SNX8 protein?

The expression and purification of functional recombinant mouse SNX8 protein requires optimization at several critical steps to ensure high yield and biological activity. Based on protein characteristics and established protocols for similar sorting nexins, the following parameters represent optimal conditions:

Expression System Selection:
Insect cell expression systems (Sf9 or Hi5 cells) typically provide the best balance of yield and functional quality for SNX8. These systems offer eukaryotic post-translational modifications while avoiding mammalian cell contamination concerns. The baculovirus expression vector system using the pFastBac vector with a C-terminal His6-tag generally yields functional protein.

Culture and Induction Conditions:

  • Cell density at infection: 1.5-2 × 10^6 cells/mL

  • MOI (multiplicity of infection): 1-2

  • Harvest time: 48-72 hours post-infection

  • Culture temperature: Reducing from 27°C to 24°C after infection can improve folding

  • Media supplementation: Addition of 1% fetal bovine serum can improve yield

Cell Lysis Optimization:

  • Lysis buffer: 50 mM HEPES pH 7.5, 300 mM NaCl, 10% glycerol, 1 mM DTT, 1 mM PMSF

  • Protease inhibitors: Complete EDTA-free protease inhibitor cocktail

  • Lysis method: Sonication (6 cycles of 10 seconds on/20 seconds off) or pressure homogenization

  • Clearing centrifugation: 40,000 × g for 45 minutes at 4°C

Purification Strategy:

  • Affinity chromatography (Ni-NTA for His-tagged protein):

    • Binding buffer: 50 mM HEPES pH 7.5, 300 mM NaCl, 10% glycerol, 20 mM imidazole

    • Wash buffer: Same as binding buffer with 40 mM imidazole

    • Elution buffer: Same as binding buffer with 250 mM imidazole

    • Flow rate: 1 mL/min

  • Ion exchange chromatography (MonoQ):

    • Buffer A: 50 mM HEPES pH 7.5, 50 mM NaCl, 5% glycerol, 1 mM DTT

    • Buffer B: Same as buffer A with 1 M NaCl

    • Gradient: 0-60% buffer B over 20 column volumes

    • Flow rate: 0.5 mL/min

  • Size exclusion chromatography (Superdex 200):

    • Running buffer: 20 mM HEPES pH 7.5, 150 mM NaCl, 5% glycerol, 1 mM DTT

    • Flow rate: 0.5 mL/min

Functional Validation Tests:

  • Membrane binding assay: Liposomes containing PtdIns(3)P

  • Membrane tubulation assay: Giant unilamellar vesicles (GUVs)

  • Thermal shift assay: To verify proper folding and stability

Storage Conditions:

  • Buffer: 20 mM HEPES pH 7.5, 150 mM NaCl, 10% glycerol, 1 mM DTT

  • Concentration: 1-2 mg/mL (avoid higher concentrations to prevent aggregation)

  • Storage: Flash freeze in liquid nitrogen and store at -80°C

  • Avoid freeze-thaw cycles by storing in small (50-100 μL) aliquots

These optimized conditions should yield highly pure, functional recombinant mouse SNX8 suitable for biochemical, structural, and cell-based assays.

How should researchers design and validate siRNAs or CRISPR guides for SNX8 knockdown or knockout studies?

Effective SNX8 knockdown or knockout studies require careful design and rigorous validation of the genetic manipulation tools. The following comprehensive approach ensures specific targeting while minimizing off-target effects:

siRNA Design Guidelines:

  • Target sequence selection:

    • Design 3-4 independent siRNAs targeting different exons

    • Select 19-23 nucleotide sequences with 30-60% GC content

    • Avoid sequences with >4 consecutive identical nucleotides

    • Target regions 50-100 nucleotides downstream of the start codon and upstream of the stop codon

    • Verify uniqueness using BLAST to minimize off-target effects

  • siRNA validation metrics:

    • Confirm knockdown efficiency at both mRNA level (qRT-PCR) and protein level (Western blot)

    • Require at least 70-80% knockdown for functional studies

    • Test dose-response relationship (typically 5-50 nM final concentration)

    • Assess knockdown duration (typically 48-96 hours)

    • Include both non-targeting control siRNA and mock transfection controls

CRISPR Guide RNA Design:

  • Target selection strategy:

    • Design 3-4 gRNAs targeting early exons (preferably exon 1-3)

    • Prioritize guides with high on-target and low off-target scores using algorithms like CRISPOR or CHOPCHOP

    • Target constitutively expressed exons present in all splice variants

    • Consider the reading frame to ensure frameshift mutations disrupt protein function

    • For SNX8-specific targeting in mice, avoid regions with high homology to other sorting nexins, particularly SNX2

  • Validation requirements:

    • Genomic verification: T7 endonuclease assay, TIDE analysis, or direct sequencing of the target locus

    • Protein elimination: Western blot confirmation using validated antibodies

    • Clonal isolation: For complete knockout studies, establish and characterize single-cell-derived clones

    • Genetic complementation: Rescue experiments with exogenous SNX8 resistant to the gRNA

Essential Controls:

  • Specificity controls:

    • Phenotype rescue with RNAi-resistant or CRISPR-resistant SNX8 expression

    • Assessment of related sorting nexin expression levels to detect compensatory changes

    • Off-target analysis using whole-genome sequencing for critical studies

  • Functional validation:

    • Test for expected phenotypes based on known SNX8 functions:
      a. Lysosome morphology and function (LysoTracker staining, lysosomal enzyme assays)
      b. Virus-triggered innate immune responses
      c. IFNγ-triggered gene induction

    • Compare phenotypes between siRNA knockdown and CRISPR knockout to distinguish acute versus chronic effects

This systematic approach to designing and validating genetic tools for SNX8 manipulation ensures that observed phenotypes can be confidently attributed to specific loss of SNX8 function rather than off-target effects or compensatory mechanisms.

What is the optimal experimental design for investigating SNX8's role in viral immunity using recombinant protein?

Investigating SNX8's role in viral immunity using recombinant protein requires a multifaceted experimental design that addresses both biochemical mechanisms and functional outcomes. Based on established findings that SNX8 positively regulates RNA virus-triggered innate immune responses , the following experimental design provides a comprehensive framework:

Phase 1: Biochemical Interaction Analysis

  • VISA/MAVS interaction characterization:

    • In vitro binding assays using purified recombinant SNX8 and VISA/MAVS proteins

    • Map interaction domains through truncation and point mutation studies

    • Determine binding affinity using surface plasmon resonance or isothermal titration calorimetry

    • Assess whether binding is direct or requires additional factors

  • Aggregation dynamics assessment:

    • Reconstitute VISA/MAVS aggregation in vitro with and without recombinant SNX8

    • Use dynamic light scattering to measure aggregation kinetics

    • Employ electron microscopy to visualize aggregate structures

    • Test if pre-aggregated VISA/MAVS can still interact with SNX8

Phase 2: Cellular Reconstitution Studies

  • Rescue experiments in SNX8-deficient cells:

    • Generate SNX8-knockout cell lines (RAW264.7 macrophages or BMDMs)

    • Transfect or transduce with wild-type or mutant recombinant SNX8

    • Challenge with RNA viruses (VSV, SeV) or RNA mimetics (poly(I:C))

    • Measure downstream signaling through phospho-IRF3, NF-κB activation

    • Quantify antiviral gene induction (IFN-β, ISG15, CXCL10) by qRT-PCR and ELISA

  • Structure-function relationship studies:

    • Create domain deletion and point mutation variants of recombinant SNX8

    • Assess which domains are required for VISA/MAVS binding versus aggregation promotion

    • Determine if membrane-binding capacity (PX domain) is essential for immune function

    • Identify critical residues that specifically affect immune function without disrupting trafficking roles

Phase 3: Ex Vivo Systems Analysis

  • Primary cell validation:

    • Isolate primary immune cells (macrophages, dendritic cells) from WT and Snx8−/− mice

    • Reconstitute with recombinant SNX8 protein using protein transduction methods

    • Assess restoration of RNA virus sensing and response pathways

    • Compare effectiveness of different recombinant SNX8 variants

  • Virus replication and spread analysis:

    • Infect reconstituted cells with reporter RNA viruses (e.g., GFP-expressing VSV)

    • Measure virus replication kinetics and viral load

    • Assess cell-to-cell spread in mixed cultures

    • Determine if recombinant SNX8 introduction can restrict viral replication

Phase 4: In Vivo Translational Assessment

  • Therapeutic potential analysis:

    • Develop formulation for delivery of recombinant SNX8 protein in vivo

    • Administer to Snx8−/− mice prior to or during RNA virus infection

    • Assess viral loads, inflammatory markers, and survival outcomes

    • Compare effectiveness of prophylactic versus therapeutic administration

MeasurementTechniqueExpected Outcome in WTExpected Outcome in SNX8-deficientExpected Outcome with Rescue
VISA/MAVS aggregationNative gel, confocal microscopyRobust aggregation after viral challengeImpaired aggregationRestored aggregation
IRF3 phosphorylationWestern blot, flow cytometryStrong inductionReduced inductionDose-dependent restoration
IFN-β productionqRT-PCR, ELISAHigh levels after stimulationSignificantly reducedRestored to near-WT levels
Viral replicationPlaque assay, qPCRControlled replicationEnhanced replicationReduced viral load
Survival rateKaplan-Meier analysisHigh survivalReduced survivalImproved survival

This experimental design comprehensively addresses both the mechanistic aspects of SNX8's role in viral immunity and its potential therapeutic applications, providing a thorough investigation framework.

How can researchers differentiate between SNX8's trafficking functions and immune signaling roles in experimental systems?

Differentiating between SNX8's membrane trafficking functions and its immune signaling roles presents a significant experimental challenge, as these functions may be interconnected or utilize shared domains. A systematic approach employing domain-specific mutations, compartment-specific targeting, and temporal analysis can effectively distinguish these roles:

1. Domain-Specific Mutational Analysis:

Creating a panel of SNX8 variants with mutations targeting specific functional domains enables precise dissection of role-specific requirements:

Domain/RegionFunctionMutation StrategyExpected Effect on TraffickingExpected Effect on Immune Signaling
PX DomainPhosphoinositide bindingR43A, R44A mutationsDisrupted endosomal localizationMay affect immune signaling if membrane localization is required
BAR DomainMembrane curvature sensing/inductionDeletion or critical residue mutationsImpaired tubulation abilityMay not affect protein-protein interactions with signaling components
Tyr95, Tyr126JAK1-mediated phosphorylation sitesY95F, Y126F mutationsLikely minimal effectAbolished IFNγ-triggered signaling
VISA/MAVS binding regionImmune complex formationAlanine scanning mutationsPotentially minimal effectDisrupted antiviral response

These variants should be introduced into SNX8-knockout cells, followed by parallel assessment of trafficking functions (lysosomal morphology, cargo sorting) and immune responses (virus-induced or IFNγ-triggered signaling).

2. Subcellular Targeting Approach:

Forcing SNX8 localization to specific compartments can reveal location-dependent functions:

  • Mitochondrial-targeted SNX8: Adding a mitochondrial targeting sequence should maintain proximity to VISA/MAVS (which localizes to mitochondria) while disrupting endosomal trafficking functions

  • Plasma membrane-targeted SNX8: Adding a plasma membrane targeting domain should prevent both endosomal function and mitochondrial signaling

  • Nuclear-targeted SNX8: Adding a nuclear localization signal should abolish both membrane trafficking and cytoplasmic signaling functions

Comparing the rescue capacity of these targeted variants for trafficking versus immune phenotypes can reveal compartment-specific requirements.

3. Temporal Separation Strategy:

Exploiting the different temporal dynamics of trafficking versus signaling events:

  • Acute inhibition: Using auxin-inducible degron (AID) system for rapid SNX8 depletion can reveal immediate effects (likely signaling) versus delayed effects (potentially trafficking-dependent)

  • Pulse-chase analysis: Following the kinetics of both processes after stimulation can separate immediate signaling events from downstream trafficking-dependent processes

  • Synchronized stimulation: Using temperature-sensitive vesicular stomatitis virus (ts-VSV) allows synchronized infection and precise temporal analysis of subsequent events

4. Cargo-Specific Functional Assays:

Developing parallel assays that specifically measure distinct SNX8 functions:

  • Trafficking function: Monitor lysosome tubulation under starvation conditions, retromer-dependent cargo sorting, or specific toxin transport

  • Immune signaling: Measure VISA/MAVS aggregation, IRF3 phosphorylation, IFNγ-induced gene expression

Importantly, these assays should be performed in the same cellular background with the same SNX8 variants to enable direct comparisons.

5. Interactome Analysis:

Comparing SNX8's protein interaction networks under different conditions:

  • Baseline versus immune-stimulated conditions (viral infection or IFNγ treatment)

  • Membrane fraction versus cytosolic fraction interactome

  • Temporal analysis of interaction dynamics during signaling activation

By systematically applying these approaches, researchers can effectively distinguish between SNX8's trafficking and signaling functions, potentially revealing whether these roles are truly separable or represent different aspects of an integrated biological function.

What are the most promising therapeutic applications of recombinant SNX8 based on current research findings?

Based on current research findings, recombinant SNX8 shows considerable therapeutic potential in several disease areas where its molecular functions directly address underlying pathological mechanisms:

  • Lysosomal Storage Disorders (LSDs):
    The most well-established therapeutic application stems from SNX8's ability to promote lysosome tubulation and reformation. Research has demonstrated that SNX8 overexpression successfully rescued LSD phenotypes in both cellular and mouse models . Specifically, AAV-based delivery of SNX8 to the brain rescued key pathological features in a Sandhoff disease mouse model (Hexb−/−), including reduction of GM2 ganglioside storage and prevention of neuronal loss . This represents a particularly promising approach because:

    • It addresses a fundamental process (lysosome reformation) affected in multiple LSDs

    • It potentially offers a complementary strategy to existing enzyme replacement therapies

    • It demonstrated efficacy in a mammalian disease model with behavioral improvements

  • Viral Infections:
    SNX8's established role as a positive regulator of the RNA virus-triggered innate immune response suggests therapeutic potential against viral pathogens . Snx8−/− mice show increased susceptibility to RNA virus infections, indicating that SNX8 augmentation could enhance antiviral defense. Therapeutic approaches might include:

    • Temporary enhancement of SNX8 expression or activity during acute viral infections

    • SNX8-derived peptides that specifically enhance VISA/MAVS aggregation

    • Small molecules that mimic SNX8's effect on antiviral signaling pathways

  • Bacterial Infections:
    Research has shown that SNX8-deficient mice infected with Listeria monocytogenes exhibited higher bacterial loads and increased mortality . This suggests SNX8-based therapeutics could enhance innate immunity against intracellular bacterial pathogens through its role in IFNγ-triggered signaling pathways.

  • Neurodegenerative Diseases:
    Given SNX8's function in maintaining lysosomal homeostasis and its successful application in neurological LSDs, there's potential relevance to other neurodegenerative conditions characterized by protein aggregation and impaired cellular clearance, such as Alzheimer's and Parkinson's diseases.

For therapeutic development, several delivery approaches show promise:

Delivery MethodAdvantagesChallengesMost Suitable Applications
AAV-mediated gene therapyLong-term expression, neurotropic serotypes availableImmunogenicity, packaging size limitations, production complexityCNS-focused LSDs, chronic conditions
Recombinant protein with cell-penetrating peptidesDirect activity control, transient effect, potential for localized deliveryProtein stability, tissue penetration, manufacturing costAcute infections, localized applications
Small molecule modulators of SNX8 activityOral bioavailability potential, lower cost, tunable pharmacokineticsTarget specificity, identification challengesWidespread applications if developed

The most immediate and promising therapeutic application remains in LSDs, where proof-of-concept has already been established in animal models. The development of optimized delivery methods and thorough safety assessment represent the next critical steps toward clinical translation.

How might the study of SNX8 interactions with novel binding partners lead to new biological insights?

The study of SNX8 interactions with novel binding partners represents a fertile ground for biological discovery, potentially revealing unexpected connections between membrane trafficking, cellular signaling, and disease pathways. Several strategic approaches can maximize the insights gained from investigating these interactions:

  • Systems-wide Interaction Mapping:
    Employing unbiased proteomics approaches such as BioID, APEX proximity labeling, or IP-MS under various cellular conditions (starvation, viral infection, IFNγ stimulation) can reveal condition-specific interaction networks. These comprehensive interactomes may uncover:

    • Connections between endosomal trafficking and immune signaling pathways

    • Novel regulatory mechanisms for lysosome reformation

    • Unexpected roles in cellular stress responses

    • Potential disease-relevant interactions

  • Protein Complex Structural Studies:
    Structural analysis of SNX8 in complex with its binding partners (JAK1, IKKβ, VISA/MAVS) could reveal:

    • Conformational changes upon binding that explain activation mechanisms

    • Critical interface residues that could be targeted therapeutically

    • The molecular basis for phosphorylation-dependent interactions

    • How a single protein can participate in seemingly diverse cellular processes

  • Cross-pathway Integration Analysis:
    SNX8's established roles in both trafficking and signaling suggest it may function as a molecular hub integrating multiple cellular pathways. Investigating this possibility could reveal:

    • How membrane trafficking events influence immune signaling efficiency

    • Whether SNX8 acts as a scaffold for multiple signaling complexes

    • If SNX8-mediated trafficking regulates the availability of signaling components

    • Whether cellular metabolic state affects SNX8 function through its role in lysosome reformation

  • Disease-specific Interaction Alterations:
    Comparing SNX8 interactomes in healthy versus disease states could provide mechanistic insights into pathological processes:

    • In LSDs: Identifying altered interactions that explain impaired lysosome reformation

    • In viral infections: Determining if viral proteins directly target SNX8 to evade immunity

    • In neurodegenerative conditions: Revealing potential connections to protein aggregation mechanisms

  • Evolutionary Perspective:
    Comparative analysis of SNX8 interactions across species could reveal:

    • Core evolutionarily conserved functions versus species-specific adaptations

    • How immune functions might have evolved from trafficking roles

    • Whether pathogen pressure has shaped SNX8 function in different species

The integration of these approaches has significant potential to yield transformative biological insights, potentially revealing SNX8 as a key nexus in cellular homeostasis networks rather than simply a component of discrete pathways. This holistic understanding could fundamentally change our conceptualization of how membrane trafficking and signaling pathways are coordinated in health and disease.

What technological advancements would facilitate better understanding of SNX8 dynamics in live cells?

Understanding the dynamic behavior of SNX8 in live cells represents a significant challenge that requires advanced imaging and molecular tools. Several technological advancements would substantially enhance our ability to track SNX8's movements, interactions, and functional states in real-time:

  • Advanced Fluorescent Protein Technologies:

    • Split fluorescent proteins optimized for SNX8 fusion to visualize protein-protein interactions with minimal disruption

    • Photoactivatable or photoswitchable SNX8 fusions for pulse-chase imaging of protein pools

    • FRET-based SNX8 biosensors to detect conformational changes upon membrane binding or partner interactions

    • Far-red and near-infrared fluorescent protein fusions for deeper tissue imaging and reduced phototoxicity

  • Super-resolution Microscopy Enhancements:

    • Lattice light-sheet microscopy with adaptive optics for high-speed 3D tracking of SNX8-positive tubules with minimal photodamage

    • STORM/PALM imaging with multi-color capabilities to simultaneously track SNX8 and its binding partners at nanoscale resolution

    • Correlative light and electron microscopy (CLEM) workflows optimized for membrane tubulation events

    • Label-free imaging techniques such as transient absorption microscopy that don't require potentially disruptive protein tagging

  • Live-cell Biochemical Probes:

    • Fluorogenic substrates for monitoring lysosomal enzyme activity in SNX8-positive compartments

    • Membrane tension and curvature sensors to correlate with SNX8 recruitment

    • Phosphoinositide biosensors to track lipid dynamics during SNX8-mediated tubulation

    • Genetically encoded calcium indicators to correlate lysosomal calcium release with SNX8 activity

  • Genome Engineering and Optogenetic Tools:

    • CRISPR knock-in of small epitope tags or fluorescent proteins at the endogenous SNX8 locus

    • Optogenetic control of SNX8 recruitment to specific membranes to induce tubulation on demand

    • Light-inducible protein-protein interaction systems to trigger SNX8 binding to specific partners

    • Degron-based systems for acute, reversible depletion of SNX8 in live cells

  • Artificial Intelligence and Computational Approaches:

    • Machine learning algorithms for tracking complex tubular structures in noisy microscopy data

    • Deep learning models to predict membrane deformation based on SNX8 concentration and distribution

    • Automated detection and classification of SNX8-positive compartments and their maturation states

    • Integration of imaging and -omics data to correlate visual phenotypes with molecular signatures

  • In vivo Imaging Advances:

    • Tissue clearing techniques compatible with SNX8 fluorescent protein preservation

    • Miniaturized microscopes for in vivo imaging of SNX8 dynamics in freely moving animals

    • Multi-photon excitation optimized for SNX8 fluorescent fusions in deep tissue imaging

    • Transgenic mouse models with conditional fluorescent SNX8 expression in specific cell types

  • Multimodal Analysis Platforms:

    • Systems integrating live-cell imaging with mass spectrometry for direct correlation of visual dynamics with molecular composition

    • Microfluidic devices for real-time manipulation of cellular environment while imaging SNX8 dynamics

    • Combined electrophysiology and imaging setups to correlate membrane dynamics with lysosomal ion fluxes

    • High-content imaging platforms for systematic perturbation analysis of SNX8 dynamics

These technological advancements would collectively enable unprecedented insights into SNX8's spatiotemporal dynamics, revealing how it coordinates its diverse functions in membrane trafficking and signaling pathways in living cells and organisms.

What emerging technologies might revolutionize therapeutic applications of SNX8 in lysosomal storage disorders?

Emerging technologies across multiple disciplines hold tremendous potential to revolutionize SNX8-based therapeutic approaches for lysosomal storage disorders (LSDs). These innovations could address current limitations in delivery, specificity, and efficacy:

  • Advanced Gene Delivery Technologies:

    • Next-generation AAV capsids: Engineered through directed evolution or machine learning to enhance blood-brain barrier penetration, reduce immunogenicity, and increase specificity for affected cell types in LSDs

    • Non-viral delivery systems: Lipid nanoparticles with tissue-targeting ligands specifically designed for SNX8 mRNA delivery with reduced immunogenicity compared to viral vectors

    • In vivo gene editing: CRISPR-based approaches to correct primary LSD mutations while simultaneously enhancing endogenous SNX8 expression through promoter engineering

    • Extracellular vesicle-based delivery: Engineered exosomes carrying SNX8 mRNA or protein for enhanced cell penetration and reduced immunogenicity

  • Protein Engineering and Delivery Innovations:

    • Cell-penetrating SNX8 variants: Fusion of cell-penetrating peptides or protein transduction domains to enable direct delivery of recombinant SNX8 protein

    • Subcellular targeting modifications: Addition of specific targeting sequences to direct SNX8 to disease-relevant compartments

    • Conditionally active SNX8: Engineered variants activated specifically in diseased lysosomes based on pH or enzyme activity differences

    • Stabilized protein formulations: Polymer encapsulation or PEGylation strategies to enhance circulation time and tissue distribution of recombinant SNX8

  • Small Molecule Approaches:

    • SNX8 activity enhancers: High-throughput screening to identify compounds that enhance endogenous SNX8 function or expression

    • Proteolysis-targeting chimeras (PROTACs): Bifunctional molecules that selectively degrade negative regulators of SNX8 activity

    • Pharmacological chaperones: Compounds that stabilize mutant lysosomal proteins complementary to SNX8's lysosome reformation activity

    • Blood-brain barrier shuttles: Conjugated delivery systems to enhance CNS penetration of SNX8-targeting therapeutics

  • Combination Therapy Platforms:

    • Dual-function vectors: Single delivery systems encoding both SNX8 and disease-specific lysosomal enzymes for synergistic treatment

    • Temporally regulated expression systems: Inducible promoters allowing pulsed SNX8 expression optimized to cellular needs

    • Cell therapy approaches: Genetically modified stem cells with enhanced SNX8 expression for cell replacement in affected tissues

    • Integrated treatment algorithms: Personalized protocols combining enzyme replacement, substrate reduction, and SNX8-based therapies based on patient-specific disease markers

  • Advanced Monitoring Technologies:

    • Real-time biomarkers: Implantable biosensors to monitor lysosomal function and adjust SNX8 therapy dosing

    • Imaging-guided therapy: MRI-visible nanoparticles co-delivered with SNX8 therapeutics to track distribution

    • Digital medicine approaches: Wearable devices integrated with SNX8 delivery systems for symptom-triggered dosing

    • AI-based progression models: Machine learning algorithms to predict optimal intervention timing based on biomarker profiles

  • Manufacturing and Scale-up Innovations:

    • Continuous bioprocessing: Advanced bioreactor technologies for cost-effective production of recombinant SNX8

    • Synthetic biology platforms: Cell-free protein synthesis systems for rapid, high-purity SNX8 production

    • 3D bioprinting: Tissue models for personalized testing of SNX8 therapeutic efficacy

    • Automated formulation technologies: Robotic systems for precision manufacturing of complex SNX8 therapeutics

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 2025 TheBiotek. All Rights Reserved.