SNX5 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on the shipping method and location. For specific delivery information, please contact your local distributor.
Synonyms
0910001N05Rik antibody; 1810032P22Rik antibody; AU019504 antibody; D2Ertd52e antibody; FLJ10931 antibody; OTTHUMP00000030340 antibody; OTTHUMP00000062927 antibody; OTTMUSP00000003367 antibody; OTTMUSP00000003368 antibody; RP11-504H3.2 antibody; RP23-35E16.2 antibody; snoRNA MBI-43 antibody; SNX 5 antibody; SNX5 antibody; SNX5_HUMAN antibody; Sorting nexin 5 antibody; Sorting nexin-5 antibody
Target Names
Uniprot No.

Target Background

Function
SNX5 plays a crucial role in various stages of intracellular trafficking. It interacts with membranes containing phosphatidylinositol 3-phosphate (PtdIns(3P)) or phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2). SNX5 functions, in part, as a component of the retromer membrane-deforming SNX-BAR subcomplex. The SNX-BAR retromer facilitates retrograde transport of cargo proteins from endosomes to the trans-Golgi network (TGN) and participates in endosome-to-plasma membrane transport for cargo protein recycling. The SNX-BAR subcomplex likely deforms the donor membrane into a tubular profile known as an endosome-to-TGN transport carrier (ETC). While SNX5 itself does not exhibit in vitro vesicle-to-membrane remodeling activity, it is involved in retrograde transport of the lysosomal enzyme receptor IGF2R. Furthermore, SNX5 potentially serves as a link between endosomal transport vesicles and dynactin. Evidence suggests that SNX5 plays a role in the internalization of EGFR following EGF stimulation. It is involved in EGFR endosomal sorting and degradation, a process potentially dependent on PIP5K1C isoform 3 and independent of the retromer. In conjunction with PIP5K1C isoform 3, SNX5 aids in HGS interaction with ubiquitinated EGFR, initiating EGFR sorting to intraluminal vesicles (ILVs) of the multivesicular body for subsequent lysosomal degradation. SNX5 also participates in E-cadherin sorting and degradation, specifically inhibiting PIP5K1C isoform 3-mediated E-cadherin degradation. Additionally, SNX5 plays a role in macropinocytosis.
Gene References Into Functions
  1. Elevated SNX5 expression is linked to alphavirus infections. PMID: 29743363
  2. Sorting nexin 5 (SNX5), a constituent of the retromer and a component of the retrograde transport pathway, interacts with UL35 proteins. The binding of UL35 proteins to SNX5 is essential for efficient viral replication and for the transport of the most abundant cytomegalovirus glycoprotein B to the cytoplasmic viral assembly center. PMID: 29444945
  3. The Chlamydia trachomatis IncE binding site is unique to human SNX5 and its related family members, SNX6 and SNX32. PMID: 28226239
  4. Sorting nexin 5 and dopamine D1 receptor regulate the expression of the insulin receptor in human renal proximal tubule cells PMID: 25825816
  5. Internalization of agonist-activated D1R is regulated by both SNX5 and GRK4, and SNX5 is critical for the recycling of the receptor to the plasma membrane PMID: 23195037
  6. Research indicates that sorting nexin 5 (SNX5) induces caspase-2 in papillary thyroid carcinoma PMID: 22486813
  7. SNX5 is localized to a subdomain of the early endosomes and is recruited to the cell membrane following EGF stimulation. PMID: 15561769
  8. SNX5 and SNX6 may serve as functional equivalents of Vps17p in the mammalian retromer. PMID: 17148574
  9. DOCK180 regulates CI-MPR trafficking through SNX5, and this function is independent of its guanine nucleotide exchange factor activity toward Rac1 PMID: 18596235

Show More

Hide All

Database Links

HGNC: 14969

OMIM: 605937

KEGG: hsa:27131

STRING: 9606.ENSP00000366988

UniGene: Hs.316890

Protein Families
Sorting nexin family
Subcellular Location
Endosome. Early endosome. Early endosome membrane; Peripheral membrane protein; Cytoplasmic side. Cell membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasmic vesicle membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasm. Cell projection, phagocytic cup. Cell projection, ruffle.

Q&A

What is SNX5 and what role does it play in macropinocytosis?

SNX5 (Sorting Nexin 5) is a 404 amino acid protein with an observed molecular weight of 47 kDa that belongs to the sorting nexin family of cytoplasmic and membrane-associated proteins characterized by the presence of a phospholipid-binding PX domain . SNX5 functions as a critical regulator of macropinocytosis, particularly in primary macrophages. Research demonstrates that SNX5 is recruited to newly-formed macropinosomes (fluid-filled endocytic vesicles >500 nm in diameter) where it influences the uptake and processing of soluble antigens . Depletion studies show that SNX5 reduction dramatically decreases the uptake of labeled dextran, a marker of macropinocytosis, indicating that SNX5 is essential for efficient macropinocytosis rather than simply associated with the process . Unlike its related family member SNX1, SNX5 appears capable of functioning independently in this capacity, as SNX1 knockout does not affect macropinosome biogenesis .

How is SNX5 structurally associated with cellular compartments?

SNX5 exhibits distinct localization patterns in cellular systems, primarily residing in endosomal compartments where it participates in sorting and transport of membrane proteins . Immunofluorescence studies reveal that endogenous SNX5 is recruited to the boundary of macropinosomes where it juxtaposes with EEA1 (Early Endosome Antigen 1), an early endosome marker also found on newly-formed macropinosomes . This positioning is critical for SNX5's function in vesicular trafficking. While SNX5 can form heterodimers with SNX1, as demonstrated through co-immunoprecipitation experiments in primary macrophages, the functional independence of SNX5 suggests that it possesses unique structural properties that allow it to modulate macropinocytosis through mechanisms distinct from those of other sorting nexins .

In which tissues and cell types is SNX5 expression most prominent?

SNX5 displays a diverse tissue distribution pattern that underscores its importance in various physiological processes. Positive Western blot detection has been reported in multiple human cell lines including HEK-293T, K-562, Jurkat, and U2OS cells, as well as in mouse cell lines such as Neuro-2a . At the tissue level, SNX5 has been detected in both human and mouse kidney tissue, as well as mouse brain tissue through immunohistochemistry . The expression of SNX5 appears to be regulated during cellular differentiation, with studies noting that SNX5 expression is lower in hematopoietic stem cells compared to committed progenitor cells . This differential expression pattern suggests that SNX5 may play specialized roles in fully differentiated cells, particularly those that rely heavily on endocytic processes such as macrophages .

What are the optimal dilution ratios for different applications of FITC-conjugated SNX5 antibody?

When working with FITC-conjugated SNX5 antibody, optimal dilution ratios must be established for each experimental application to ensure specific signal while minimizing background fluorescence. Based on available data for unconjugated SNX5 antibodies that would apply similarly to FITC conjugates, the following dilution ranges are recommended as starting points:

ApplicationRecommended Dilution Range
Immunofluorescence1:100-1:400
Flow Cytometry1:50-1:200
Confocal Microscopy1:100-1:400
High-Content Imaging1:100-1:200

It is critically important to note that these dilutions should be titrated in each testing system to obtain optimal results, as sample type and fixation methods can significantly impact antibody performance . For immunofluorescence applications specifically, a stepwise titration approach beginning with 1:100 dilution and conducting parallel experiments at 1:200, 1:300, and 1:400 is recommended to identify the optimal signal-to-noise ratio for your specific cell type and fixation protocol.

How can researchers effectively visualize SNX5 in macropinocytosis studies using FITC conjugates?

To effectively visualize SNX5 in macropinocytosis studies using FITC-conjugated antibodies, researchers should implement a dual-labeling approach that captures both the dynamic process of macropinocytosis and SNX5 localization. A methodologically sound approach includes:

  • Prime macrophages or relevant cells by stimulation with appropriate factors (e.g., CSF-1 at 50 ng/ml for macrophages) .

  • Label newly-formed macropinosomes using Texas Red-conjugated 70 kDa dextran (or similar fluid-phase marker) by incubating cells at 37°C for 15 minutes .

  • Fix cells using 4% paraformaldehyde while preserving membrane structure.

  • If using indirect immunofluorescence, permeabilize cells with 0.1% Triton X-100 and block with appropriate serum.

  • For direct visualization with FITC-conjugated SNX5 antibody, apply at optimized dilution and incubate for 1-2 hours at room temperature or overnight at 4°C.

  • Counter-stain with DAPI to visualize nuclei.

  • Mount slides using anti-fade mounting medium to preserve FITC fluorescence.

  • Analyze using confocal microscopy, identifying macropinosomes as circular structures >500 nm in diameter positive for dextran .

This approach allows simultaneous visualization of macropinosomes (red) and SNX5 (green), providing clear evidence of SNX5 recruitment to macropinosome membranes and enabling quantitative analysis of colocalization.

What fixation and permeabilization protocols are optimal for preserving FITC signal in SNX5 antibody experiments?

Preserving both antigen integrity and FITC fluorescence requires carefully optimized fixation and permeabilization protocols. For SNX5 detection using FITC-conjugated antibodies, the following methodology is recommended:

  • Fixation options:

    • For membrane-associated proteins like SNX5, 4% paraformaldehyde in PBS for 15 minutes at room temperature preserves membrane structure while maintaining antigen accessibility.

    • Alternatively, methanol fixation (-20°C for 10 minutes) may improve detection of some epitopes but can diminish FITC fluorescence intensity.

  • Permeabilization considerations:

    • For paraformaldehyde-fixed samples, 0.1-0.2% Triton X-100 in PBS for 5-10 minutes provides sufficient membrane permeabilization without excessive protein extraction.

    • Saponin (0.1%) offers gentler permeabilization and may better preserve membrane-associated structures.

  • Antigen retrieval:

    • For tissue sections or challenging samples, antigen retrieval with TE buffer at pH 9.0 has been demonstrated to enhance SNX5 detection .

    • Alternatively, citrate buffer at pH 6.0 may be employed if alkaline conditions yield suboptimal results .

  • Post-fixation treatment:

    • A quenching step using 50 mM NH₄Cl for 10 minutes can reduce autofluorescence from aldehyde fixation.

    • Thorough blocking with 5-10% normal serum matching the secondary antibody host (if using indirect detection) minimizes non-specific binding.

These protocols should be systematically tested and optimized for each experimental system, as fixation requirements may vary between different cell types and tissue preparations.

How can researchers address weak or inconsistent FITC-SNX5 antibody signals?

When confronted with weak or inconsistent signals using FITC-conjugated SNX5 antibody, a systematic troubleshooting approach should be implemented:

  • Antibody titration and storage:

    • Confirm antibody concentration through spectrophotometric measurement if signal appears consistently weak.

    • Verify storage conditions - FITC conjugates should be stored at -20°C, protected from light, and maintained in buffer containing 50% glycerol for stability .

    • Avoid repeated freeze-thaw cycles that can degrade both antibody function and FITC fluorescence.

  • Sample preparation optimization:

    • Review fixation protocol - overfixation can mask epitopes while underfixation may result in sample degradation.

    • Ensure complete permeabilization while avoiding excessive membrane disruption.

    • For tissues requiring antigen retrieval, compare TE buffer pH 9.0 and citrate buffer pH 6.0 to determine optimal retrieval conditions .

  • Signal amplification strategies:

    • Implement tyramide signal amplification if direct FITC conjugates provide insufficient signal.

    • Consider longer incubation times (overnight at 4°C) to improve antibody penetration in complex samples.

    • Utilize high-sensitivity detection systems such as confocal microscopy with photomultiplier optimization.

  • Expression level considerations:

    • Be aware that SNX5 expression varies by cell type, with lower expression in hematopoietic stem cells compared to differentiated cells .

    • Confirm target protein expression in your sample using parallel Western blot analysis.

  • Photobleaching prevention:

    • Use anti-fade mounting media containing anti-photobleaching agents.

    • Minimize exposure to excitation light during microscopy sessions.

    • Acquire images rapidly using high-sensitivity cameras to reduce exposure times.

These systematic approaches address the most common causes of weak signals when working with FITC-conjugated antibodies against SNX5.

What controls should be included when using FITC-conjugated SNX5 antibody in microscopy experiments?

Rigorous experimental design requires appropriate controls to validate findings with FITC-conjugated SNX5 antibody. The following controls should be included:

  • Specificity controls:

    • Positive control: Include a cell line or tissue with confirmed high SNX5 expression (e.g., HEK-293T, mouse kidney tissue) .

    • Negative control: Utilize SNX5 knockdown/knockout samples generated through siRNA or CRISPR-Cas9 techniques to confirm signal specificity .

    • Peptide competition assay: Pre-incubate antibody with excess immunizing peptide to confirm epitope-specific binding.

  • Technical controls:

    • Isotype control: Include a FITC-conjugated isotype-matched irrelevant antibody (e.g., FITC-conjugated rabbit or mouse IgG) to assess non-specific binding.

    • Autofluorescence control: Examine unstained samples to identify intrinsic cellular fluorescence.

    • Single-color controls: For multi-color experiments, include single-stained samples to establish proper compensation settings.

  • Functional validation:

    • Colocalization control: Confirm SNX5 localization pattern through colocalization with established markers such as EEA1 for early endosomes .

    • Physiological response control: Verify expected changes in SNX5 distribution following stimulation (e.g., CSF-1 treatment in macrophages) .

  • Quantification controls:

    • Acquisition control: Maintain identical microscopy settings (exposure, gain, offset) across all experimental conditions.

    • Background subtraction control: Include regions lacking cells to establish background fluorescence levels.

These comprehensive controls ensure that observations are attributable to specific SNX5 detection rather than technical artifacts or non-specific binding.

How can researchers distinguish between specific and non-specific binding of FITC-conjugated SNX5 antibody?

Distinguishing specific from non-specific binding is crucial for generating reliable data with FITC-conjugated SNX5 antibody. Implement these methodological approaches:

  • Concentration-dependent binding assessment:

    • Perform a dilution series (1:50, 1:100, 1:200, 1:400) of FITC-conjugated SNX5 antibody.

    • Specific binding will show consistent localization patterns despite dilution, while non-specific binding typically diminishes at higher dilutions.

    • Plot signal-to-noise ratio against antibody concentration to identify optimal working dilution.

  • Pattern recognition analysis:

    • Compare observed staining patterns with established SNX5 localization - expected to show endosomal/macropinosomal membrane association and cytoplasmic distribution .

    • Non-specific binding typically presents as diffuse background, nuclear staining, or edge artifacts.

    • Verify subcellular localization by co-staining with established markers (e.g., EEA1 for early endosomes) .

  • Genetic validation:

    • Compare staining patterns between wild-type cells and those with SNX5 depletion (50-90% reduction achieved through miRNA techniques has been documented) .

    • Quantify signal reduction in SNX5-depleted cells - specific binding should show proportional reduction with protein level.

  • Competitive inhibition:

    • Pre-incubate cells with excess unconjugated SNX5 antibody before applying FITC-conjugated version.

    • Specific binding sites should be saturated, resulting in significantly reduced FITC signal.

  • Multiple antibody validation:

    • Compare staining patterns from different antibody clones targeting distinct SNX5 epitopes.

    • Concordant patterns from independent antibodies strongly support specificity.

These approaches provide multiple lines of evidence to distinguish genuine SNX5 detection from artifacts that could confound experimental interpretation.

How can FITC-conjugated SNX5 antibody be utilized to study the interaction between SNX5 and SNX1?

Investigating the SNX5-SNX1 interaction requires sophisticated methodological approaches that leverage the properties of FITC-conjugated SNX5 antibody. The following protocol can be implemented:

  • Co-immunoprecipitation validation:

    • Prior to fluorescence studies, confirm SNX5-SNX1 interaction biochemically using co-immunoprecipitation as established in primary macrophages .

    • Immunoprecipitate endogenous SNX5 using specific antibodies and blot for SNX1 to confirm the 70 kDa band representing the interacting partner .

  • Dual immunofluorescence approach:

    • Utilize FITC-conjugated SNX5 antibody alongside a spectrally distinct (e.g., Cy3 or Alexa 594) SNX1 antibody.

    • Apply sequential staining protocol to prevent potential steric hindrance between antibodies.

    • Image using confocal microscopy with appropriate filter sets to minimize spectral overlap.

  • Live-cell interaction dynamics:

    • For cells amenable to transfection, combine FITC-SNX5 antibody detection with expression of fluorescently-tagged SNX1 (e.g., mCherry-SNX1).

    • Implement live-cell antibody feeding techniques with membrane-permeable FITC-SNX5 conjugates.

    • Perform time-lapse imaging to capture dynamic association during macropinosome formation.

  • Proximity ligation assay enhancement:

    • Combine FITC-conjugated SNX5 antibody with unconjugated SNX1 antibody in a modified proximity ligation assay.

    • Visualize protein interactions (<40 nm proximity) as discrete fluorescent spots while simultaneously observing SNX5 distribution through direct FITC fluorescence.

  • Quantitative interaction analysis:

    • Measure colocalization using Pearson's or Mander's coefficients to quantify the spatial relationship between SNX5 and SNX1.

    • Implement FRET (Fluorescence Resonance Energy Transfer) analysis if using appropriate fluorophore combinations to confirm direct molecular interaction.

This multifaceted approach provides both visualization and quantification of the SNX5-SNX1 interaction in cellular contexts, building upon the established biochemical evidence .

What methods can be used to study the role of SNX5 in antigen processing using FITC-conjugated antibodies?

To investigate SNX5's role in antigen processing using FITC-conjugated SNX5 antibody, researchers should implement this comprehensive methodological framework:

  • Macrophage preparation and manipulation:

    • Isolate primary macrophages (peritoneal or bone marrow-derived) following established protocols .

    • Generate SNX5-depleted macrophages using verified miRNA constructs (e.g., miRNA-1.4) that achieve ~89% reduction in SNX5 levels .

    • Confirm SNX5 depletion through Western blot and immunofluorescence using FITC-conjugated SNX5 antibody.

  • Antigen uptake and processing assessment:

    • Incubate macrophages with fluorescently-labeled model antigens (e.g., DQ-Ovalbumin which becomes fluorescent upon proteolytic processing).

    • Track the kinetics of antigen uptake and processing in control versus SNX5-depleted cells.

    • Co-stain with FITC-conjugated SNX5 antibody to correlate processing efficiency with SNX5 expression levels.

  • Macropinosome formation and maturation:

    • Label newly-formed macropinosomes using fluorescent dextran (70 kDa) as established in previous studies .

    • Track macropinosome maturation through co-staining with endosomal/lysosomal markers.

    • Quantify macropinosome formation rates and relate to antigen processing efficiency.

  • Functional readouts of antigen presentation:

    • Assess surface presentation of processed antigens using flow cytometry.

    • Measure T-cell activation in co-culture systems to determine functional consequences of SNX5 depletion.

    • Correlate SNX5 expression levels (quantified by FITC-SNX5 staining intensity) with antigen presentation capacity.

  • Mechanistic dissection:

    • Investigate whether SNX5 functions independently or requires SNX1 in antigen processing contexts.

    • Utilize SNX1 knockout macrophages to determine if SNX5 localization and function in antigen processing remain intact .

This approach enables comprehensive analysis of SNX5's role in the antigen processing pathway, building on established findings that SNX5 depletion dramatically reduces soluble antigen uptake and processing in macrophages .

How can researchers quantitatively analyze changes in SNX5 distribution during cellular processes?

Quantitative analysis of SNX5 distribution dynamics requires rigorous methodological approaches that maximize the utility of FITC-conjugated SNX5 antibody:

  • Automated high-content imaging protocol:

    • Perform time-course experiments capturing SNX5 distribution at defined intervals following stimulation.

    • Implement automated image acquisition across multiple fields to generate statistically robust datasets.

    • Apply consistent thresholding algorithms to segment cellular regions and identify SNX5-positive structures.

  • Morphometric analysis parameters:

    • Quantify number, size, intensity, and subcellular distribution of SNX5-positive structures.

    • Measure SNX5 recruitment to macropinosomes as ratio of membrane-associated versus cytosolic signal.

    • Track changes in SNX5 distribution patterns during macropinosome maturation or following pharmacological interventions.

  • Colocalization quantification:

    • Calculate Pearson's correlation coefficient between SNX5 and markers of different endocytic compartments.

    • Implement object-based colocalization analysis to determine percentage of SNX5-positive structures containing specific markers.

    • Generate distance maps to assess spatial relationships between SNX5 and other trafficking components.

  • Dynamic redistribution metrics:

    • For live-cell applications, calculate rates of SNX5 recruitment to newly forming macropinosomes.

    • Measure residence time of SNX5 on macropinosomes during maturation.

    • Quantify SNX5 membrane/cytosol partitioning coefficients under different experimental conditions.

  • Statistical analysis framework:

    • Apply appropriate statistical tests to determine significance of observed changes.

    • Implement analysis of variance for multi-condition experiments.

    • Generate distribution plots rather than simple averages to capture heterogeneity in cellular responses.

This comprehensive quantitative approach transforms descriptive observations into robust metrics that can reveal subtle phenotypes and temporal dynamics in SNX5 function across diverse experimental contexts.

How can FITC-conjugated SNX5 antibody be incorporated into multi-parameter flow cytometry experiments?

Incorporating FITC-conjugated SNX5 antibody into multi-parameter flow cytometry requires careful experimental design and optimization:

  • Panel design considerations:

    • Position FITC-SNX5 antibody appropriately within your panel considering that FITC emits at 519nm and may have spillover into PE channels.

    • Pair with compatible fluorophores to minimize compensation requirements (e.g., APC, PE-Cy7, BV421).

    • Account for relative abundance of SNX5 (typically moderate expression) when selecting fluorophore brightness.

  • Intracellular staining protocol:

    • Optimize fixation (4% paraformaldehyde, 10 minutes) and permeabilization (0.1% saponin or commercial permeabilization buffer).

    • Include protein transport inhibitors if examining dynamics following stimulation.

    • Implement sequential surface marker staining before fixation and permeabilization for SNX5 detection.

  • Validation controls:

    • Include FITC-conjugated isotype control matched to the SNX5 antibody.

    • Prepare SNX5-depleted control samples (e.g., using verified siRNA approaches) .

    • Generate compensation controls for all fluorophores in the panel.

  • Analysis strategy:

    • Gate on viable, single cells before analyzing SNX5 expression.

    • Create ratio parameters to examine relative SNX5 levels between experimental conditions.

    • Consider dimensionality reduction techniques (tSNE, UMAP) for complex datasets examining SNX5 in relation to multiple markers.

  • Functional correlation:

    • Implement intracellular cytokine staining to correlate SNX5 levels with functional outputs.

    • Include phagocytic/endocytic capacity measurements (fluorescent bead uptake) to relate SNX5 expression to function.

    • Sort cells based on SNX5 expression levels for subsequent functional assays.

This methodological framework enables integration of SNX5 analysis into complex immunophenotyping experiments while maintaining rigorous standards for specificity and quantitative accuracy.

What approaches can researchers use to study the dynamics of SNX5 using FITC-conjugated antibodies in live cells?

Studying SNX5 dynamics in live cells using FITC-conjugated antibodies requires specialized techniques that preserve cell viability while enabling specific labeling:

  • Cell-permeable antibody delivery:

    • Conjugate FITC-SNX5 antibody with cell-penetrating peptides (e.g., TAT sequence) to facilitate membrane penetration.

    • Alternatively, employ reversible permeabilization techniques such as digitonin treatment at low concentrations.

    • Utilize antibody electroporation in specialized cell types that maintain viability.

  • Minimally invasive introduction methods:

    • Implement microinjection of FITC-SNX5 antibody for precise delivery to individual cells.

    • Employ microfluidic cell squeezing techniques that create transient pores for antibody entry.

    • Consider glass bead loading for adherent cells to introduce antibodies without compromised viability.

  • Complementary approaches:

    • Combine FITC-SNX5 antibody with expression of spectrally distinct fluorescent protein-tagged markers of endocytic compartments.

    • Utilize photoactivatable or photoconvertible FITC derivatives to perform pulse-chase experiments tracking SNX5 movements.

    • Implement FRAP (Fluorescence Recovery After Photobleaching) to measure SNX5 dynamics at specific cellular locations.

  • Advanced imaging configurations:

    • Utilize spinning disk confocal microscopy for rapid acquisition with minimal phototoxicity.

    • Implement light sheet microscopy for extended imaging sessions with reduced photodamage.

    • Consider lattice light sheet microscopy for high-resolution 3D dynamics of SNX5-positive structures.

  • Environmental controls:

    • Maintain physiological temperature, pH, and CO₂ levels throughout imaging.

    • Minimize phototoxicity through reduced exposure times and optimized illumination strategies.

    • Include antioxidants in imaging media to mitigate photobleaching effects on FITC.

These specialized techniques enable examination of SNX5 dynamics in physiologically relevant contexts while overcoming the traditional limitations of antibody applications in live-cell imaging.

How can researchers effectively perform quantitative comparison of SNX5 expression levels across different tissues or experimental conditions?

Quantitative comparison of SNX5 expression levels requires standardized methodologies that ensure reproducibility and accuracy:

This comprehensive approach enables reliable quantitative comparisons of SNX5 expression levels across diverse experimental contexts, from cultured cell lines to primary tissues and disease models, while maintaining rigorous standards for data quality and reproducibility.

How might FITC-conjugated SNX5 antibody facilitate investigation of SNX5's role in disease models?

FITC-conjugated SNX5 antibody provides versatile capabilities for investigating SNX5's role in various disease models through these methodological approaches:

  • Neurodegenerative disorder models:

    • Track SNX5 distribution in neuronal models of Alzheimer's or Parkinson's disease.

    • Correlate SNX5 levels with endosomal dysfunction phenotypes using high-resolution confocal microscopy.

    • Investigate receptor trafficking defects that may contribute to pathology using pulse-chase approaches with FITC-SNX5 antibody.

  • Cancer research applications:

    • Quantify SNX5 expression across tumor types using tissue microarrays and automated image analysis.

    • Correlate SNX5 levels with growth factor receptor trafficking alterations in cancer cells.

    • Investigate SNX5 as a potential biomarker through multiplexed immunofluorescence panels.

  • Infectious disease models:

    • Track SNX5 recruitment to pathogen-containing compartments using co-localization analysis.

    • Investigate potential pathogen interference with SNX5-dependent trafficking pathways.

    • Utilize SNX5 depletion in conjunction with FITC-SNX5 antibody to assess functional consequences on pathogen clearance.

  • Inflammatory conditions:

    • Examine SNX5 dynamics in macrophages during inflammatory activation.

    • Correlate SNX5 levels with altered antigen presentation in autoimmune disease models.

    • Investigate potential therapeutic targeting of SNX5-dependent pathways.

  • Development of advanced analytical tools:

    • Implement machine learning algorithms for automated detection of altered SNX5 distribution patterns.

    • Develop computational models of SNX5 trafficking dynamics based on quantitative imaging data.

    • Generate predictive frameworks linking SNX5 expression patterns to disease progression.

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.