RAB39A Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Structure and Function

The RAB39A antibody (HPA042505) is a polyclonal antibody derived from rabbit serum. It is affinity-purified using the immunogen sequence SQRQVTREEAEKLSADCGMKYIETSA, ensuring specificity for the RAB39A protein. Key characteristics include:

AttributeDetails
TargetHuman RAB39A protein (UniProt accession pending)
Immunogen SequenceSQRQVTREEAEKLSADCGMKYIETSA
ReactivityHuman tissues (validated via IHC across 44 normal and 20 cancer types)
Dilution Range1:1000–1:2500 for IHC (buffered aqueous glycerol solution)
ApplicationImmunohistochemistry, cancer stem cell studies, and lysosomal trafficking

Immunohistochemistry

The antibody is validated for IHC across diverse tissue types, including cancerous and normal specimens. It localizes RAB39A to lysosomal compartments and areas of active autophagy, enabling studies on phagosome maturation .

Cancer Stemness and Tumorigenesis

Research employing this antibody has demonstrated RAB39A's role in maintaining cancer stem cell (CSC) populations. For example, silencing RAB39A in osteosarcoma cells reduces spherogenicity and tumorigenesis, while overexpression enhances these traits .

Role in Phagosome Maturation

  • Antigen Cross-Presentation: RAB39A facilitates the conversion of phagosomes into MHC-I peptide-loading compartments, enhancing CD8+ T-cell responses .

  • Autophagy Regulation: In macrophages, RAB39A interacts with PI3K to suppress LPS-induced autophagy, modulating inflammatory responses .

Cancer-Associated Functions

  • RAB39A-RXRB Axis: Studies reveal RAB39A complexes with RXRB (Retinoid X Receptor Beta) to regulate CSC self-renewal. Silencing RAB39A downregulates RXRB, impairing tumorigenesis .

  • Lysosomal Dynamics: RAB39A promotes lysosomal acidification, a critical factor in cancer stemness and drug resistance .

Table 2: RAB39A Functional Interactions

PartnerRole
MHC-I MoleculesPromotes peptide loading in phagosomes
PI3KRegulates autophagy in macrophages
RXRBMediates cancer stemness via transcriptional regulation

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery information.
Synonyms
rab related GTP binding protein antibody; Rab-39 antibody; Rab39 antibody; RAB39A antibody; RAB39A member RAS oncogene family antibody; Ras related protein Rab39A antibody; Ras-related protein Rab-39A antibody; RB39A_HUMAN antibody
Target Names
RAB39A
Uniprot No.

Target Background

Function
RAB39A plays a crucial role in the maturation and acidification of phagosomes that engulf pathogens, such as Staphylococcus aureus and Mycobacterium tuberculosis. It is involved in vesicular trafficking and the fusion of phagosomes with lysosomes. RAB39A negatively regulates lipopolysaccharide (LPS)-induced autophagosome formation in macrophages, potentially by implicating phosphatidylinositol 3-kinase (PI3K). It may also participate in multiple neurite formation.
Gene References Into Functions
  1. Molecular pathways regulated by RAB39A are transcriptionally maintained through the formation of a molecular complex with retinoid X receptor beta (RXRB), nuclear receptor corepressor (NCOR), and histone deacetylase (HDAC), which also contribute to cancer stemness. PMID: 29648608
  2. C9ORF72, a gene associated with amyotrophic lateral sclerosis (ALS), causes suboptimal autophagy. PMID: 27494456
  3. Research indicates that Rab39a promotes chlamydial replication and infectivity. This study is the first to demonstrate the involvement of a late endocytic Rab GTPase in chlamydial infection development. PMID: 26163492
  4. Results show that Rab39a interacts with caspase-1 and suggest that Rab39a acts as a trafficking adaptor, connecting caspase-1 to interleukin-1 beta (IL-1beta) secretion. PMID: 19833722
Database Links

HGNC: 16521

KEGG: hsa:54734

STRING: 9606.ENSP00000322594

UniGene: Hs.354906

Protein Families
Small GTPase superfamily, Rab family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side. Cytoplasmic vesicle, phagosome. Cytoplasmic vesicle, phagosome membrane; Lipid-anchor; Cytoplasmic side. Lysosome. Note=Recruited to phagosomes containing S.aureus or M.tuberculosis.

Q&A

What is RAB39A and why is it significant in immunological research?

RAB39A is a member of the Rab family of small GTPases that plays a critical role in vesicular trafficking and phagosome maturation. It has emerged as a key regulator of antigen cross-presentation (XPT) by dendritic cells, a process essential for CD8+ T cell responses against cancer and viral infections. RAB39A specifically promotes the delivery of MHC-I molecules from the endoplasmic reticulum to phagosomes and increases levels of peptide-empty MHC-I conformers that can be loaded with peptide in this compartment . Unlike other trafficking proteins, RAB39A functions selectively in the cross-presentation pathway but does not affect MHC-II presentation or the classical MHC-I pathway, making it a valuable target for studies focused on understanding the mechanics of cross-presentation .

Which cell types express RAB39A and how can this expression be detected?

RAB39A shows differential expression across immune cell populations. Studies using RAB39A knockout mice with LacZ-containing constructs revealed that expression is highest in CD11c-positive dendritic cells, particularly in the CD8α+ subset, which expressed the highest levels of RAB39A . CD11b+CD11c+ DCs expressed intermediate amounts, while a fraction of B220-positive cells (B lymphocytes) and CD11b-positive cells (macrophages) showed lower expression levels . For detection, fluorescein-di-beta-D-galactopyranoside (FDG) has been used as a β-galactosidase substrate in knockout models to identify cells that would normally express RAB39A . When selecting antibodies for detecting native RAB39A expression, consider using flow cytometry for quantitative analysis of expression levels across different immune cell subsets.

What are the recommended protocols for immunofluorescence staining of RAB39A in dendritic cells?

For optimal immunofluorescence staining of RAB39A in dendritic cells, consider the following methodological approach:

  • Isolate dendritic cells from mouse spleen or differentiate bone marrow-derived dendritic cells according to standard protocols

  • Adhere cells to poly-L-lysine coated coverslips for 30-60 minutes

  • Fix cells with 4% paraformaldehyde for 15 minutes at room temperature

  • Permeabilize with 0.1% Triton X-100 for 5 minutes

  • Block with 5% normal serum corresponding to the secondary antibody species for 1 hour

  • Incubate with anti-RAB39A primary antibody (1:100-1:500 dilution range) overnight at 4°C

  • Wash 3 times with PBS

  • Incubate with fluorophore-conjugated secondary antibody for 1 hour at room temperature

  • Counterstain with DAPI for nuclear visualization

  • Mount with anti-fade mounting medium

For co-localization studies, consider dual staining with markers such as MHC-I (using antibodies like H2-Kb clone AF6-88.5), phagosomal markers like CD107a/b, or other trafficking components like Sec22b that have been shown to interact with RAB39A in cross-presentation pathways .

How should I validate the specificity of a RAB39A antibody?

Validating RAB39A antibody specificity requires multiple complementary approaches:

  • Genetic knockout controls: Compare staining between wild-type and RAB39A knockout cells/tissues. The complete absence of signal in knockout samples strongly supports antibody specificity .

  • siRNA knockdown: Similar to knockout validation but using transient silencing. Reduced signal intensity proportional to knockdown efficiency further confirms specificity.

  • Overexpression systems: Cells transfected with RAB39A expression constructs should show increased signal compared to non-transfected controls. The doxycycline-inducible systems used in RAB39A research provide excellent positive controls .

  • Western blot analysis: Verify that the antibody detects a single band of the expected molecular weight (~25 kDa for RAB39A).

  • Peptide competition: Pre-incubation of the antibody with the immunizing peptide should eliminate or significantly reduce specific staining.

These validation steps are essential before proceeding with experimental applications to ensure reliable and reproducible results.

How can I distinguish between GDP-bound and GTP-bound forms of RAB39A?

Differentiating between the GDP-bound (inactive) and GTP-bound (active) forms of RAB39A requires specialized techniques, as standard antibodies typically cannot distinguish between these conformational states. Research has shown that proper cycling between GDP- and GTP-bound forms is essential for RAB39A function in cross-presentation, as neither GDP-locked nor GTP-locked mutants could enhance cross-presentation of bead-bound antigen . To study these distinct forms:

  • Pull-down assays: Use GST-fusion proteins containing the binding domains of RAB39A effectors that specifically bind to the GTP-bound form. This approach has been established for many Rab proteins.

  • Mutant expression: Generate and express RAB39A mutants that are either GDP-locked (typically S22N mutation) or GTP-locked (typically Q72L mutation) and compare their localization and function to wild-type RAB39A .

  • Proximity ligation assays: Detect interactions between RAB39A and known effectors that only bind to the GTP-bound form, providing spatial information about where active RAB39A is localized.

These approaches require careful experimental design and appropriate controls, including comparison to the wild-type RAB39A that can cycle between GDP and GTP forms, which has been shown to be necessary for proper function in cross-presentation .

What methodologies can be used to study RAB39A's role in phagosomal maturation?

To investigate RAB39A's role in phagosomal maturation and its function in converting phagosomes into MHC-I peptide-loading compartments, consider these methodological approaches:

  • Phagosome isolation and proteomic analysis:

    • Feed cells latex beads coated with antigens (like Ova)

    • Isolate phagosomes by density gradient centrifugation

    • Compare protein composition between phagosomes from wild-type and RAB39A-deficient cells by mass spectrometry to identify RAB39A-dependent changes in phagosome composition

  • Live cell imaging of phagosome dynamics:

    • Express fluorescently-tagged RAB39A (e.g., GFP-RAB39A)

    • Track phagosome maturation in real-time using live confocal microscopy

    • Measure co-localization with markers of early endosomes, late endosomes, and lysosomes

  • Functional assessment of phagosomal activity:

    • Measure phagosomal pH using pH-sensitive dyes or ratiometric probes

    • Assess proteolytic activity within phagosomes using fluorogenic substrates

    • Determine levels of reactive oxygen species (ROS) using appropriate indicators, as RAB39A has been shown to increase NOX2 levels and ROS production in phagosomes

  • Analysis of MHC-I loading in phagosomes:

    • Use antibodies specific for peptide-empty MHC-I conformers (similar to the H2-Ld open conformer antibody 64-3-7 mentioned in the materials)

    • Assess the presence of peptide-loaded MHC-I complexes using conformation-specific antibodies

These techniques can help elucidate how RAB39A modifies phagosomes to become peptide-loading compartments capable of efficient cross-presentation.

How can I effectively use RAB39A antibodies for co-immunoprecipitation studies?

For successful co-immunoprecipitation (co-IP) of RAB39A and its interaction partners, follow these methodological recommendations:

  • Cell lysis optimization:

    • Use mild lysis buffers containing 1% NP-40 or 1% digitonin to preserve protein-protein interactions

    • Include protease inhibitors and phosphatase inhibitors to prevent degradation

    • For GTPases like RAB39A, consider adding GTP-γS (a non-hydrolyzable GTP analog) to stabilize interactions with effector proteins

  • Antibody selection and validation:

    • Test multiple RAB39A antibodies for immunoprecipitation efficiency

    • Validate specificity using RAB39A knockout or knockdown controls

    • Consider using epitope-tagged RAB39A (e.g., HA-tagged as used in the research) if native antibodies perform poorly

  • Experimental procedure:

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

    • Incubate with anti-RAB39A antibody overnight at 4°C

    • Add protein A/G beads and incubate for 2-4 hours

    • Wash extensively with lysis buffer containing reduced detergent

    • Elute bound proteins and analyze by immunoblotting

  • Controls to include:

    • IgG control from the same species as the RAB39A antibody

    • Lysate from RAB39A-deficient cells

    • Input samples (5-10% of the lysate used for IP)

    • Reverse co-IP using antibodies against suspected interaction partners

This approach has successfully identified RAB39A-interacting proteins involved in cross-presentation, including Sec22b and components of the NOX2 complex .

What experimental designs can best demonstrate RAB39A's specific role in cross-presentation?

To effectively demonstrate RAB39A's specific role in cross-presentation while ruling out effects on other antigen presentation pathways, design comprehensive experiments that compare multiple presentation pathways within the same experimental system. Based on published research approaches , consider the following design:

  • Cell models:

    • Use dendritic cell lines with inducible or knockdown RAB39A expression

    • Include wild-type, RAB39A-overexpressing, and RAB39A-deficient conditions

    • Consider primary dendritic cells from both wild-type and RAB39A knockout mice

  • Antigen presentation pathways to compare:

    • Cross-presentation pathway: Exogenous antigens presented on MHC-I (e.g., OVA-coated beads)

    • Classical MHC-I pathway: Endogenous expression of antigens (e.g., cytosolic OVA)

    • MHC-II pathway: Exogenous antigens presented on MHC-II molecules

  • Readout systems:

    • Use reporter T cells specific for each pathway (e.g., B3Z for OVA peptide/H-2Kb, MF2-Luc for I-Ab-OVA)

    • Measure antigen-specific T cell activation via IL-2 production, luciferase activity, or proliferation

    • Perform dose-response experiments across a range of antigen concentrations

  • Controls:

    • Pathway-specific controls: β2M siRNA to inhibit MHC-I presentation and H2-I-Ab siRNA to block MHC-II presentation

    • Antigen processing controls: Compare processed peptides vs. whole proteins

    • Construct a results table similar to this format:

Antigen Presentation PathwayEffect of RAB39A KnockdownEffect of RAB39A Overexpression
Cross-presentation (P2C)Decreased presentationEnhanced presentation
Classical MHC-I pathwayNo significant effectNo significant effect
MHC-II presentationNo significant effectNo significant effect

This comprehensive approach will highlight RAB39A's selective role in the cross-presentation pathway, as demonstrated in the research findings .

How should I optimize immunoblotting protocols for detecting RAB39A in different cellular compartments?

For optimal detection of RAB39A in different cellular compartments by immunoblotting, consider these methodological recommendations:

  • Sample preparation by subcellular fractionation:

    • Prepare whole cell lysates as a reference sample

    • Isolate cytosolic, membrane, and organelle fractions using differential centrifugation

    • For phagosome isolation, use latex bead-containing phagosomes purified on sucrose gradients

    • Include compartment-specific markers as controls (e.g., calnexin for ER, LAMP1 for lysosomes)

  • Protein extraction optimization:

    • For membrane-associated proteins like RAB39A, use lysis buffers containing 0.5-1% NP-40 or Triton X-100

    • Include protease inhibitors to prevent degradation

    • Sonicate briefly to enhance extraction from membrane compartments

  • SDS-PAGE and transfer conditions:

    • Use 12-15% polyacrylamide gels for optimal resolution of small GTPases (~25 kDa)

    • Transfer to PVDF membranes at lower voltage (30V) overnight at 4°C for efficient transfer of small proteins

    • Verify transfer efficiency using reversible protein stains before blocking

  • Immunodetection:

    • Block with 5% non-fat dry milk or BSA in TBST

    • Incubate with anti-RAB39A antibody (1:500-1:2000 dilution range)

    • Include loading controls appropriate for each subcellular fraction

    • Use HRP-conjugated secondary antibodies and enhanced chemiluminescence detection

  • Validation steps:

    • Include RAB39A knockout/knockdown samples as negative controls

    • For overexpression studies, use doxycycline-inducible systems as positive controls

    • Validate fractionation purity using compartment-specific markers

This optimized protocol will allow reliable detection of RAB39A across different cellular compartments, facilitating studies of its trafficking and localization during cross-presentation.

What controls are essential when studying RAB39A's interaction with NOX2 and Sec22b?

When investigating RAB39A's interactions with NOX2 and Sec22b, which have been shown to be critical for its function in cross-presentation , include these essential controls:

  • Genetic controls:

    • RAB39A knockout/knockdown cells

    • NOX2-deficient cells (to establish specificity of the interaction)

    • Sec22b-depleted cells (to determine if the interaction is direct or indirect)

    • Cells expressing GDP-locked or GTP-locked RAB39A mutants (to determine if interactions are nucleotide-dependent)

  • Biochemical controls for co-immunoprecipitation experiments:

    • IgG isotype control antibodies

    • Reverse co-IP (immunoprecipitate with anti-NOX2 or anti-Sec22b and blot for RAB39A)

    • Competition with excess recombinant proteins

    • Treatment with GDP or non-hydrolyzable GTP analogs to manipulate RAB39A conformation

  • Imaging controls for co-localization studies:

    • Secondary antibody-only controls to rule out non-specific staining

    • Non-overlapping fluorophores with minimal spectral overlap

    • Quantitative co-localization analysis (Pearson's or Mander's coefficients)

    • Z-stack acquisition to confirm true co-localization in three dimensions

  • Functional readouts to validate biological significance:

    • ROS production measurements (as RAB39A increases NOX2-dependent ROS)

    • Phagosomal pH measurements (as RAB39A promotes phagosome alkalinization)

    • Cross-presentation assays with model antigens

    • Measurement of MHC-I loading in phagosomes

What are common pitfalls when using RAB39A antibodies in immunohistochemistry and flow cytometry?

When using RAB39A antibodies for immunohistochemistry and flow cytometry, researchers should be aware of these common pitfalls and their solutions:

  • High background signal:

    • Cause: Insufficient blocking, excessive antibody concentration, or non-specific binding

    • Solution: Optimize blocking conditions (try 5-10% serum or BSA), titrate antibody concentration, include 0.1-0.3% Triton X-100 in blocking buffer

  • Variable expression detection across cell types:

    • Cause: Different fixation sensitivities or epitope accessibility in various cell populations

    • Solution: Compare multiple fixation methods (paraformaldehyde, methanol, acetone) and validate with RAB39A knockout controls in each cell type

  • Limited detection in tissue sections:

    • Cause: Poor antibody penetration or epitope masking during fixation

    • Solution: Optimize antigen retrieval methods (heat-induced or enzymatic), extend primary antibody incubation time, use thinner tissue sections

  • False positives in flow cytometry:

    • Cause: Autofluorescence, particularly in phagocytic cells

    • Solution: Include FMO (fluorescence minus one) controls, use spectral flow cytometry, and employ autofluorescence quenching protocols

  • Inconsistent staining between experiments:

    • Cause: Antibody lot variation or instability

    • Solution: Purchase larger lots when possible, aliquot antibodies to avoid freeze-thaw cycles, validate each new lot against previous ones

  • False negatives in RAB39A-expressing cells:

    • Cause: Low expression levels in certain cell types or epitope masking

    • Solution: Use signal amplification methods (tyramide signal amplification, polymer detection systems) or consider alternative detection approaches like the FDG substrate method used for detecting RAB39a-expressing cells in knockout models

Careful optimization and inclusion of appropriate controls will help ensure reliable detection of RAB39A across different experimental systems.

How can I distinguish between specific roles of RAB39A and other vesicular trafficking proteins in cross-presentation?

Differentiating the specific contributions of RAB39A from other vesicular trafficking proteins in cross-presentation requires thoughtful experimental design:

  • Combinatorial genetic manipulation:

    • Generate single knockouts/knockdowns for RAB39A and other trafficking proteins (e.g., Sec22b, Rab27a, Rab11)

    • Create double knockouts/knockdowns to identify synergistic or redundant functions

    • Analyze phenotypes using cross-presentation assays with reporter T cells

  • Pathway-specific readouts:

    • Measure distinct steps in the cross-presentation pathway, including:

      • Antigen uptake and phagosome formation

      • Phagosomal pH regulation

      • ROS production (specifically RAB39A-enhanced)

      • Antigen export to cytosol

      • MHC-I recruitment to phagosomes

      • Peptide loading onto MHC-I molecules

  • Rescue experiments with domain-specific mutants:

    • Express wild-type RAB39A or specific mutants (GDP-locked, GTP-locked) in knockout cells

    • Create chimeric proteins containing domains from RAB39A and other Rab proteins

    • Assess which domains confer specificity for cross-presentation

  • Temporal analysis of protein recruitment:

    • Use live-cell imaging with fluorescently tagged proteins

    • Determine the temporal sequence of different trafficking proteins during phagosome maturation

    • Identify rate-limiting steps regulated by RAB39A versus other factors

  • Cell type-specific analysis:

    • Compare the requirement for RAB39A in cross-presentation across different DC subsets

    • Correlate with expression levels of other trafficking proteins

    • As demonstrated in research, RAB39A's contribution varies between CD8α+ and CD8α- dendritic cells

These approaches will help delineate the unique contribution of RAB39A to the cross-presentation pathway and its functional relationships with other vesicular trafficking proteins.

What analytical methods can be used to quantify RAB39A-dependent changes in phagosome composition?

To quantitatively assess RAB39A-dependent changes in phagosome composition, researchers can employ these analytical methods:

  • Quantitative proteomics of isolated phagosomes:

    • Isolate phagosomes from wild-type and RAB39A-deficient cells

    • Perform stable isotope labeling (SILAC) or tandem mass tag (TMT) labeling

    • Analyze by liquid chromatography-tandem mass spectrometry (LC-MS/MS)

    • Compare relative protein abundances to identify RAB39A-dependent changes

    • Focus on proteins like MHC-I, Sec22b, and NOX2 components which are known to be affected by RAB39A

  • Flow cytometry of isolated phagosomes:

    • Isolate latex bead-containing phagosomes

    • Stain with fluorescently labeled antibodies against proteins of interest

    • Analyze by flow cytometry for quantitative assessment of protein levels

    • Compare phagosome populations from different conditions

  • Microscopy-based quantification:

    • Perform immunofluorescence staining of phagosomes in intact cells

    • Acquire high-resolution confocal z-stacks

    • Use automated image analysis to quantify:

      • Signal intensity of proteins of interest

      • Co-localization coefficients between markers

      • Phagosome size and morphology parameters

  • Biochemical activity assays:

    • Measure specific enzymatic activities in isolated phagosomes:

      • ROS production (using luminol or DCF-DA)

      • Proteolytic activity (using fluorogenic substrates)

      • pH (using ratiometric dyes)

    • Compare activities between RAB39A-positive and RAB39A-negative phagosomes

  • Single-phagosome analysis:

    • Track individual phagosomes over time using live-cell imaging

    • Quantify recruitment kinetics of fluorescently tagged proteins

    • Correlate with functional outcomes like antigen cross-presentation

These quantitative approaches can reveal both the composition and functional properties of phagosomes that are specifically regulated by RAB39A, providing insight into its mechanism of action in promoting cross-presentation.

How can I resolve contradictory results when studying RAB39A in different dendritic cell subsets?

When facing contradictory results in RAB39A studies across different dendritic cell subsets, consider these methodological approaches to resolve discrepancies:

  • Standardize isolation and culture conditions:

    • Use consistent methods for isolating DC subsets (e.g., magnetic separation, flow sorting)

    • Standardize culture conditions, including media, supplements, and activation stimuli

    • Control for maturation state, as DC maturation can alter cross-presentation machinery

  • Account for expression level differences:

    • Quantify RAB39A expression levels across DC subsets (as research shows highest expression in CD8α+ DCs, intermediate in CD11b+CD11c+ DCs)

    • Normalize functional readouts to RAB39A expression levels

    • Consider the impact of expression levels on dependency (higher expressing cells may have compensatory mechanisms)

  • Examine redundant pathways:

    • Investigate alternative cross-presentation mechanisms in different DC subsets

    • Assess the relative contribution of various pathways (P2C vs. vacuolar)

    • Research has shown that CD8α+ DCs (which express highest RAB39A levels) show less dependency on RAB39A than CD8α- DCs, potentially due to redundant mechanisms

  • Consider antigen-specific variables:

    • Test multiple antigen forms (soluble, bead-bound, cell-associated)

    • Vary antigen concentration to identify threshold effects

    • Examine antigen uptake efficiency across DC subsets

  • Comprehensive statistical analysis:

    • Perform power analysis to ensure adequate sample sizes

    • Use mixed-effects models to account for inter-experimental variation

    • Conduct meta-analysis across experiments to identify consistent trends

  • Complementary approaches:

    • Compare in vitro and in vivo results to identify system-specific effects

    • Use both primary cells and cell lines to distinguish cell type-specific vs. general mechanisms

    • Employ both loss-of-function (knockout/knockdown) and gain-of-function (overexpression) approaches

By systematically addressing these factors, researchers can resolve contradictory results and develop a more nuanced understanding of RAB39A's role across different dendritic cell populations.

How can RAB39A antibodies be used to study cross-presentation defects in disease models?

RAB39A antibodies can be valuable tools for investigating cross-presentation abnormalities in disease models through these methodological approaches:

  • Tumor microenvironment studies:

    • Compare RAB39A expression and localization in tumor-infiltrating DCs vs. normal tissue DCs

    • Assess correlation between RAB39A levels and cross-presentation capacity

    • Investigate if tumor-derived factors alter RAB39A function or expression

    • Determine if restoring RAB39A levels can enhance anti-tumor immunity

  • Infectious disease models:

    • Examine whether pathogens target RAB39A to evade cross-presentation

    • Compare RAB39A recruitment to phagosomes containing pathogens vs. inert particles

    • Assess if pathogen-induced changes in phagosomal pH affect RAB39A function

    • Determine if increasing RAB39A expression enhances cross-presentation of microbial antigens

  • Autoimmune disease analysis:

    • Investigate if RAB39A polymorphisms or expression changes are associated with autoimmune disorders

    • Examine whether aberrant RAB39A function contributes to inappropriate cross-presentation of self-antigens

    • Test if modulating RAB39A activity affects autoimmune disease progression

  • Aging-related immune dysfunction:

    • Compare RAB39A expression and function in DCs from young vs. aged individuals

    • Determine if age-related changes in cross-presentation correlate with altered RAB39A activity

    • Assess whether restoring RAB39A function improves vaccine responses in aged subjects

  • Methodological approach:

    • Immunohistochemistry of tissue sections to detect RAB39A expression patterns

    • Flow cytometry to quantify RAB39A levels in specific DC subsets

    • Immunoprecipitation to assess RAB39A interactions with partner proteins

    • Functional assays to correlate RAB39A status with cross-presentation capacity

Since RAB39A is selectively involved in cross-presentation rather than other antigen presentation pathways , changes in its expression or function may provide specific insights into cross-presentation defects in various disease states.

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.