VPS18 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Composition and Development

VPS18 antibodies are typically raised in mouse (IgG2a) or rabbit (IgG) hosts, targeting specific epitopes of the 110 kDa VPS18 protein . HRP conjugation involves covalent attachment of the enzyme to the antibody via lysine residues, a process standardized in protocols like Abcam’s Lightning-Link® system . Key characteristics include:

ParameterSpecification
TargetVPS18 (UniProt: Q9P253)
Host SpeciesMouse (Clone 2E10G11) or Rabbit (Polyclonal)
ConjugateHRP (direct detection)
ReactivityHuman, Rat, Mouse
ApplicationsWB, IHC, IF/ICC, ELISA
Storage-20°C in PBS with 0.02% sodium azide and 50% glycerol

Vesicular Trafficking and Lysosomal Function

VPS18 is a core component of the HOPS/CORVET complexes, which mediate endosome-lysosome fusion . HRP-conjugated VPS18 antibodies have been used to:

  • Track VPS18 localization in A549, HeLa, and HepG2 cells via IF/ICC, revealing its association with late endosomes/lysosomes .

  • Validate VPS18 knockdown in CRISPR screens, where its absence disrupted phagosome membrane integrity in Mycobacterium tuberculosis-infected cells .

Cancer Immunotherapy

Recent studies highlight VPS18’s role in stabilizing PD-L1, a checkpoint protein in cancer immune evasion:

  • VPS18 knockdown reduced PD-L1 levels in lung adenocarcinoma (LLC) and prostate cancer (RM1) models, enhancing T-cell cytotoxicity .

  • HRP-based detection confirmed VPS18’s interaction with VPS35, a retromer complex protein, which promotes PD-L1 glycosylation and stability .

Recommended Dilutions

ApplicationDilution RangeObserved MW (kDa)Key Tissues/Cells Tested
Western Blot1:5,000–1:50,000100–110HeLa, 293T, K562
IHC1:500–1:2,000N/AHuman liver cancer
IF/ICC1:50–1:500N/AA549 cells

Validation in Disease Models

  • Immune Checkpoint Resistance: HRP-conjugated antibodies quantified VPS18 overexpression in drug-resistant RM1/Doc prostate cancer cells, correlating with PD-L1 upregulation and reduced CD8+ T-cell infiltration .

  • Autophagy Studies: VPS18-HRP signals were critical in mapping autophagosome-lysosome fusion defects in VPS18-knockout macrophages .

Technical Considerations

  • Sensitivity: HRP conjugation reduces incubation times but may increase background in IHC; antigen retrieval with TE buffer (pH 9.0) optimizes signal .

  • Cross-Reactivity: Mouse-derived clones show stronger reactivity in human/rat samples, while rabbit polyclonals detect mouse orthologs .

  • Stability: HRP conjugates retain activity for 12 months at -20°C but degrade rapidly with freeze-thaw cycles .

Emerging Research Directions

  • Therapeutic Targeting: Small-molecule inhibitors like RDN disrupt VPS18-PD-L1 interactions, synergizing with anti-CTLA-4 therapy in metastatic cancers .

  • Neurodegeneration: Preliminary data suggest VPS18-HRP probes could map lysosomal dysfunction in models of Parkinson’s disease .

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 orders within 1-3 business days of receiving them. Delivery times may vary depending on the chosen purchasing method and location. For precise delivery estimates, kindly consult your local distributors.
Synonyms
hVPS18 antibody; PEP3 antibody; Vacuolar protein sorting 18 antibody; Vacuolar protein sorting protein 18 antibody; Vacuolar protein sorting-associated protein 18 homolog antibody; vps18 antibody; VPS18_HUMAN antibody
Target Names
VPS18
Uniprot No.

Target Background

Function
VPS18 is a protein that plays a vital role in vesicle-mediated protein trafficking to lysosomal compartments. It is involved in both endocytic membrane transport and autophagic pathways. VPS18 is believed to function as a core component of the putative HOPS and CORVET endosomal tethering complexes. These complexes are proposed to be involved in the Rab5-to-Rab7 endosome conversion, likely through the involvement of MON1A/B. VPS18 also mediates tethering and docking events during SNARE-mediated membrane fusion by binding to SNAREs and SNARE complexes. The HOPS complex is believed to be recruited to Rab7 on the late endosomal membrane, regulating late endocytic, phagocytic, and autophagic traffic towards lysosomes. The CORVET complex is thought to act as a Rab5 effector, mediating early endosome fusion, possibly within specific endosome subpopulations. VPS18 is essential for the fusion of endosomes and autophagosomes with lysosomes and plays a role in dendrite development of Purkinje cells.
Gene References Into Functions
  1. VPS18 recruits VPS41 to the human HOPS complex through a RING-RING interaction. PMID: 28931724
  2. Depletion of hVps18 or hMon2 significantly reduced the efficient production of infectious HIV-1 virions in human cells. PMID: 21450827
  3. hVPS18 may play a crucial role in regulating SNK activity through its ubiquitin ligase. PMID: 16203730
  4. Ubiquitin binding and ubiquitylation of the GGA3-GAT domain are mutually dependent on the ubiquitin ligase activity of hVPS18. PMID: 16996030
Database Links

HGNC: 15972

OMIM: 608551

KEGG: hsa:57617

STRING: 9606.ENSP00000220509

UniGene: Hs.23876

Protein Families
VPS18 family
Subcellular Location
Late endosome membrane; Peripheral membrane protein; Cytoplasmic side. Lysosome membrane; Peripheral membrane protein; Cytoplasmic side. Early endosome. Cytoplasmic vesicle, autophagosome. Cytoplasmic vesicle, clathrin-coated vesicle. Note=Cytoplasmic, peripheral membrane protein associated with early endosomes and late endosomes/lysosomes.
Tissue Specificity
Ubiquitous. Expression was highest in heart and low in lung.

Q&A

What is VPS18 and what are its primary cellular functions?

VPS18 (Vacuolar Protein Sorting-associated protein 18 homolog) is a critical component involved in vesicle-mediated protein trafficking to lysosomal compartments, participating in both endocytic membrane transport and autophagic pathways. This protein functions as a core component of two essential endosomal tethering complexes: the HOPS (homotypic fusion and protein sorting) complex and the CORVET (class C core vacuole/endosome tethering) complex . VPS18 participates in the critical Rab5-to-Rab7 endosome conversion process, which represents a key maturation step in the endocytic pathway . Through its interactions with SNAREs and SNARE complexes, VPS18 mediates tethering and docking events during membrane fusion processes that are essential for proper cellular homeostasis . Recent research has further identified VPS18 as having potential E3 ubiquitin ligase activity, suggesting additional regulatory roles beyond membrane trafficking .

Why should researchers use HRP-conjugated VPS18 antibodies rather than unconjugated versions?

HRP-conjugated VPS18 antibodies provide significant advantages for researchers investigating vesicular trafficking pathways through detection methods such as Western blotting and immunohistochemistry. The primary benefit lies in the elimination of secondary antibody steps, which streamlines experimental protocols and reduces background noise through fewer incubation and washing steps . HRP's enzymatic activity provides exceptional sensitivity for detecting even low abundance proteins like VPS18, which can be crucial when examining endogenous expression levels or studying delicate cell types where protein expression may be limited . The stable conjugation between the VPS18 antibody and HRP enzyme ensures consistent and reproducible results across experiments, which is particularly important when quantifying relative protein levels or comparing samples from different experimental conditions . Additionally, the well-established nature of HRP-based detection systems means they can be readily integrated with existing laboratory workflows and imaging systems without requiring specialized equipment or detection platforms.

How does VPS18 contribute to autophagy and lysosomal degradation pathways?

VPS18 plays an essential role in both endosomal trafficking and autophagy by facilitating the fusion of autophagosomes with lysosomes, a critical terminal step in the autophagic degradation pathway. Multiple studies have demonstrated that depletion of VPS18 results in a significant inhibition of autophagosome-lysosome fusion, leading to accumulation of autophagosomes and impaired clearance of autophagic cargo . As a core component of the HOPS complex, VPS18 helps recruit this tethering machinery to Rab7-positive late endosomal/lysosomal membranes, thereby bringing these compartments into close proximity with autophagosomes to facilitate membrane fusion events . The protein functions in concert with other HOPS components including VPS11, VPS16, VPS33A, VPS39, and VPS41, all of which are required for proper fusion of both endosomes and autophagosomes with lysosomes . Interestingly, while VIPAR and VPS33B proteins were previously suggested as alternative components in these pathways, experimental evidence indicates they form a distinct complex that is not involved in either endosome-lysosome or autophagosome-lysosome fusion processes .

What are the optimal conditions for using HRP-conjugated VPS18 antibodies in Western blotting?

When utilizing HRP-conjugated VPS18 antibodies for Western blotting, researchers should optimize several key parameters to achieve strong and specific signals. Based on manufacturer recommendations and research protocols, dilution ratios typically range from 1:100 to 1:1000, with the optimal concentration determined through systematic titration experiments for each specific antibody and cell type combination . The primary blocking agent should be selected based on the antibody specifications, with 5% non-fat dry milk in TBST or 3-5% BSA being common choices that effectively reduce background while preserving specific binding . Incubation times generally range from 1-2 hours at room temperature or overnight at 4°C, with the longer, colder incubation often providing improved signal-to-noise ratios particularly for less abundant proteins like VPS18 . For enhanced detection sensitivity, researchers can employ chemiluminescent substrates with varying degrees of sensitivity depending on the expected abundance of VPS18 in their samples, with signal exposure times typically ranging from 30 seconds to 5 minutes depending on protein expression levels .

How can researchers verify the specificity of their VPS18 antibody?

Verifying antibody specificity is crucial for ensuring reliable and reproducible results in VPS18 research. A comprehensive validation strategy should include multiple complementary approaches. First, researchers should perform siRNA or shRNA-mediated knockdown of VPS18 and observe the corresponding reduction in signal intensity on Western blots compared to control samples, which provides strong evidence for antibody specificity . Overexpression systems using tagged VPS18 constructs (such as HA-tagged VPS18) represent another valuable validation method, where detection with both the VPS18 antibody and an antibody against the tag should yield matching patterns in immunoblotting or immunofluorescence applications . Immunoprecipitation followed by mass spectrometry can provide definitive identification of the immunoprecipitated protein as VPS18, as demonstrated in studies that detected peptides of VPS11, VPS16, VPS18, VPS33A, and VPS41 in HOPS complex immunoprecipitates . Additionally, testing the antibody across multiple cell lines with known differences in VPS18 expression levels can further confirm specificity, with consistent detection patterns corresponding to expected biological variations.

What conjugation methods are used to create HRP-conjugated antibodies, and what are their implications?

The conjugation of antibodies to HRP involves several sophisticated biochemical approaches, each with distinct advantages and considerations for research applications. Heterobifunctional cross-linking reagents represent a widely used method, particularly water-soluble compounds such as sulfosuccinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (sulfo-SMCC) and N-succinimidyl S-acetylthioacetate (SATA), which enable the generation of stable antibody-HRP conjugates through controlled multistep protocols . The chemistry behind these conjugations typically involves activating HRP with sulfo-SMCC to create reactive maleimide groups that can subsequently couple to sulfhydryl groups introduced into antibodies through thiolation processes . This approach offers superior control over the extent of cross-linking while minimizing unwanted polymerization of conjugated proteins, which is crucial for maintaining both antibody affinity and enzymatic activity . The strategic introduction of sulfhydryl groups is particularly advantageous because their naturally low frequency in proteins compared to amines or carboxylates limits target antibody modification, thereby increasing the probability that the resulting VPS18 antibody-HRP conjugate will retain robust antigen-binding activity .

How should researchers design experiments to study VPS18's role in endosome-lysosome fusion?

Designing experiments to investigate VPS18's function in endosome-lysosome fusion requires careful consideration of appropriate assays, controls, and readout systems. Researchers should employ fluorescent dextran-based trafficking assays, where cells are pulsed with dextran Alexa Fluor 488 to label endosomes and subsequently assessed for colocalization with lysosomal markers such as Magic Red® to quantify fusion events . Complementary approaches should include RNAi-mediated depletion of VPS18 compared with control siRNA treatments, with careful validation of knockdown efficiency through Western blotting . For comprehensive analysis, researchers should simultaneously examine other HOPS complex components (VPS11, VPS16, VPS33A, VPS39, and VPS41) to distinguish between VPS18-specific effects and general HOPS complex dysfunction . Rescue experiments involving the re-expression of wild-type VPS18 in depleted cells provide critical evidence for specificity, while the use of mutant VPS18 constructs can help identify functional domains essential for fusion activity . Quantification should employ both visual colocalization analysis and more objective methods such as fluorescence-activated cell sorting (FACS) to measure total fluorescence levels across different experimental conditions, ensuring that observed phenotypes are not due to altered marker uptake or expression .

How can HRP-conjugated VPS18 antibodies be utilized to investigate autophagy in neurodegenerative disease models?

HRP-conjugated VPS18 antibodies offer valuable tools for investigating impaired autophagy in neurodegenerative disease models, where dysfunctional vesicular trafficking represents a common pathological feature. Researchers can employ these antibodies in Western blotting to quantify VPS18 expression levels across various disease models, correlating potential alterations with autophagy markers such as LC3-II and p62, which provides insights into whether HOPS complex dysfunction contributes to disease pathogenesis . Immunohistochemistry using HRP-conjugated VPS18 antibodies enables spatial analysis of VPS18 distribution in brain tissue sections from disease models compared to controls, with particular attention to regions exhibiting protein aggregation or neurodegeneration . This approach can reveal whether VPS18 recruitment to autophagosomes or lysosomes is compromised in disease states, potentially explaining defective clearance of protein aggregates. Dual-labeling experiments combining VPS18 antibodies with markers for disease-specific protein aggregates (such as amyloid-β, tau, or α-synuclein) can determine whether VPS18-positive structures accumulate around aggregates, suggesting attempted but incomplete clearance mechanisms .

What techniques can be used to study the interaction between VPS18 and other HOPS/CORVET complex proteins?

Investigating the interactions between VPS18 and other components of the HOPS/CORVET complexes requires sophisticated protein-protein interaction methodologies. Co-immunoprecipitation using HRP-conjugated VPS18 antibodies represents a fundamental approach, where researchers can pull down VPS18 and identify associated proteins through Western blotting or mass spectrometry, as demonstrated in studies that detected VPS11, VPS16, VPS33A, and VPS41 in VPS18 immunoprecipitates . Proximity ligation assays offer an advanced in situ technique that can visualize interactions between VPS18 and other complex components within intact cells, providing spatial information about where these complexes form and function . Researchers can also employ FRET (Förster Resonance Energy Transfer) or BRET (Bioluminescence Resonance Energy Transfer) techniques using fluorescently tagged VPS18 and other complex components to monitor real-time interactions and complex formation dynamics in living cells. Yeast two-hybrid or mammalian two-hybrid systems provide complementary genetic approaches for mapping specific interaction domains between VPS18 and its binding partners, while bacterial expression systems with purified components enable biochemical characterization of direct protein-protein interactions and binding affinities .

How can researchers investigate the newly discovered E3 ubiquitin ligase activity of VPS18?

The emerging recognition of VPS18 as an E3 ubiquitin ligase opens new avenues for research requiring specialized methodologies to characterize this enzymatic function. Researchers should conduct in vitro ubiquitination assays using purified recombinant VPS18, E1 and E2 enzymes, ubiquitin, and potential substrate proteins to directly assess VPS18's capacity to catalyze ubiquitin transfer, with Western blotting using anti-ubiquitin antibodies to detect resulting ubiquitinated products . Complementary cellular approaches involve overexpressing wild-type VPS18 versus catalytically inactive mutants (identified through structure-based predictions) followed by proteomic analysis to identify differentially ubiquitinated proteins as potential substrates . Domain mapping experiments using truncated or point-mutated VPS18 constructs can help identify the catalytic domain responsible for ubiquitin ligase activity, with particular focus on conserved RING or HECT domains typically associated with E3 ligases . To establish the physiological relevance of this activity, researchers should investigate how disruption of VPS18's E3 ligase function impacts endosome-lysosome fusion and signaling pathways such as ERα signaling, which has been linked to VPS18's ubiquitination activity .

How can researchers troubleshoot high background when using HRP-conjugated VPS18 antibodies?

High background signal is a common challenge when working with HRP-conjugated antibodies that requires systematic troubleshooting approaches. Researchers should first optimize blocking conditions by testing different blocking agents (BSA, non-fat dry milk, commercial blocking solutions) at various concentrations (3-5%) and extended blocking times (1-2 hours at room temperature), as inadequate blocking represents a primary cause of non-specific binding . Dilution optimization is equally critical, with systematic testing of the VPS18 antibody at multiple dilutions ranging from 1:100 to 1:1000 to identify the optimal concentration that maximizes specific signal while minimizing background . Additional measures include extending and increasing the number of wash steps between antibody incubations, using detergent-containing wash buffers (0.1-0.3% Tween-20 in PBS or TBS) to reduce non-specific hydrophobic interactions, and ensuring that all incubation steps are performed with gentle agitation to promote uniform antibody distribution . For Western blotting applications specifically, researchers should examine the quality of protein transfer, as incomplete or uneven transfer can create artifacts, and consider using alternative membrane types (PVDF versus nitrocellulose) which may provide different signal-to-noise ratios depending on the specific antibody characteristics .

What factors might cause variability in VPS18 detection between different experimental replicates?

Variability in VPS18 detection across experimental replicates can stem from multiple sources that researchers must systematically address. Cell culture conditions represent a significant factor, as variations in cell density, passage number, and growth phase can substantially alter VPS18 expression levels, particularly given its involvement in constitutive cellular processes like membrane trafficking . Technical considerations during sample preparation are equally important, including consistent protein extraction methods, precise protein quantification, and standardized sample loading for Western blotting applications . The stability of the HRP conjugate itself warrants attention, as repeated freeze-thaw cycles can diminish enzymatic activity, while improper storage conditions or expired reagents may lead to reduced signal intensity over time . Experimental variables during detection procedures, such as inconsistent incubation times, temperature fluctuations, or variations in substrate development conditions, can introduce significant replicate-to-replicate variability even with identical samples . To minimize these sources of variation, researchers should implement rigorous standardization protocols including consistent cell culture practices, preparation of fresh working dilutions of antibodies for each experiment, precise timing of all incubation steps, and the inclusion of appropriate housekeeping protein controls for normalization across replicates .

How can researchers interpret changes in VPS18 localization patterns across different cell types or conditions?

Interpreting variations in VPS18 localization patterns requires careful consideration of both biological and technical factors. Researchers should recognize that cell type-specific differences in endosomal-lysosomal system organization can naturally lead to distinct VPS18 distribution patterns, as demonstrated by studies showing cell-specific requirements for endosome-lysosome fusion machinery . When analyzing altered VPS18 localization under experimental conditions, it's essential to distinguish between changes in protein abundance versus redistribution, which can be accomplished by comparing Western blot data (total protein levels) with immunofluorescence patterns (spatial distribution) . Colocalization analysis with established markers for different endosomal compartments provides crucial context, including Rab5 for early endosomes, Rab7 for late endosomes, and LAMP1 for lysosomes, helping determine whether VPS18 recruitment to specific compartments is enhanced or impaired . Changes in VPS18 localization should be interpreted in the context of functional outcomes, such as altered endosome-lysosome fusion efficiency or autophagosome accumulation, as localization changes without functional consequences may represent compensatory adaptations rather than primary defects .

What are the emerging roles of VPS18 beyond its canonical functions in endosomal trafficking?

Beyond its well-established functions in endosomal trafficking, VPS18 exhibits emerging roles across diverse cellular processes that represent exciting frontiers for research. Recent studies have implicated VPS18 in dendrite development of Purkinje cells, suggesting specialized functions in neuronal morphogenesis and circuit formation that extend beyond general membrane trafficking pathways . The newly discovered E3 ubiquitin ligase activity of VPS18 opens an entirely new functional dimension, with evidence suggesting involvement in the regulation of ERα signaling pathways, potentially connecting endosomal trafficking machinery with nuclear receptor signaling networks . Some research suggests VPS18 may play a role in controlling the levels and localization of the serine/threonine-specific protein kinase AKT2 on endosomes, potentially contributing to the regulation of cellular signaling pathways beyond trafficking . The involvement of VPS18 in multiple protein complexes beyond HOPS and CORVET suggests it may function as a multifaceted regulatory node that integrates various cellular processes through its interactions with diverse protein partners .

How can multi-omics approaches incorporate VPS18 antibodies to understand vesicular trafficking networks?

Multi-omics approaches incorporating VPS18 antibodies offer powerful strategies for comprehensively mapping vesicular trafficking networks and their dysfunction in disease states. Researchers can employ immunoprecipitation with HRP-conjugated VPS18 antibodies followed by mass spectrometry (IP-MS) to identify the complete interactome of VPS18 across different cellular conditions, revealing both constitutive binding partners and condition-specific interactions that may regulate trafficking dynamics . Integration with phosphoproteomics enables identification of phosphorylation sites on VPS18 and its interacting partners, providing insights into how post-translational modifications regulate HOPS/CORVET complex assembly and function across different cellular states . Spatial proteomics approaches using VPS18 antibodies for immunoisolation of specific vesicular compartments followed by proteomic analysis can determine the protein composition of VPS18-positive structures under various experimental conditions, revealing cargo-specific trafficking pathways . Complementary transcriptomic analysis examining gene expression changes following VPS18 depletion or overexpression can identify compensatory pathways and regulatory networks connected to VPS18 function, while integration with genome-wide CRISPR screens can uncover synthetic lethal interactions that reveal functional redundancies in trafficking pathways .

What methodological advances are enabling new applications of VPS18 antibodies in live-cell imaging?

Innovative methodological advances are expanding the applications of VPS18 antibodies in live-cell imaging, providing unprecedented insights into vesicular trafficking dynamics. The development of cell-permeable nanobodies derived from VPS18 antibodies represents a significant breakthrough, allowing intracellular labeling of endogenous VPS18 in living cells without genetic modification, which enables visualization of native protein dynamics without overexpression artifacts . Single-molecule tracking approaches using quantum dot-conjugated VPS18 antibody fragments can reveal the kinetics of VPS18 recruitment to vesicular compartments at nanometer resolution, providing detailed information about complex assembly processes and residence times on different membrane compartments . Correlative light and electron microscopy (CLEM) applications utilizing HRP-conjugated VPS18 antibodies enable precise ultrastructural localization of VPS18-positive compartments, where the HRP activity can generate electron-dense reaction products visible by electron microscopy while correlating with fluorescent signals from the same structures . For monitoring dynamic processes, split-peroxidase complementation systems are being developed where one fragment is fused to a VPS18 binding partner and another to a nanobody recognizing VPS18, enabling visualization of protein-protein interactions through HRP activity only when the complex assembles .

How do applications of VPS18 antibodies differ between Western blotting, immunohistochemistry, and flow cytometry?

The applications of VPS18 antibodies across different experimental platforms involve distinct considerations for optimal results and data interpretation. In Western blotting applications, HRP-conjugated VPS18 antibodies primarily detect denatured forms of the protein, making them ideal for quantifying total VPS18 expression levels, assessing molecular weight changes due to post-translational modifications, or validating knockdown efficiency, with recommended dilutions typically ranging from 1:100-1000 . For immunohistochemistry (IHC), these antibodies recognize epitopes that remain accessible in fixed tissues, allowing spatial analysis of VPS18 distribution across different cell types and subcellular compartments, though more stringent dilutions (1:100-500) are generally required to minimize background staining in tissue sections . Flow cytometry applications using VPS18 antibodies enable quantitative assessment of protein expression across large cell populations, particularly valuable for analyzing heterogeneity in trafficking pathway components, though this requires cell permeabilization protocols optimized to maintain cellular integrity while allowing antibody access to intracellular VPS18 .

What experimental approaches can differentiate between the roles of VPS18 in HOPS versus CORVET complexes?

Distinguishing the specific functions of VPS18 within HOPS versus CORVET complexes requires sophisticated experimental strategies that selectively target complex-specific components or interactions. Researchers can employ co-immunoprecipitation with antibodies against complex-specific components—specifically VPS39 or VPS41 for HOPS and VPS3 or VPS8 for CORVET—followed by VPS18 detection to determine the relative distribution of VPS18 between these complexes under different cellular conditions . Complementary approaches include proximity ligation assays using VPS18 antibodies paired with antibodies against complex-specific components to visualize and quantify the association of VPS18 with each complex in situ, providing spatial information about where these interactions predominantly occur . Functional discrimination can be achieved through selective depletion of complex-specific components (VPS39/VPS41 for HOPS or VPS3/VPS8 for CORVET) followed by analysis of early endosome fusion (CORVET-dependent) versus late endosome-lysosome fusion (HOPS-dependent) to determine which processes are impaired when VPS18 function is compromised in each complex specifically .

ComplexSpecific ComponentsAssociated RabPrimary FunctionDetection Method
HOPSVPS39, VPS41Rab7Late endosome-lysosome fusionCo-IP with VPS41/VPS39 antibodies
CORVETVPS3, VPS8Rab5Early endosome fusionCo-IP with VPS3/VPS8 antibodies
BothVPS11, VPS16, VPS18, VPS33A-Core tethering machineryDirect IP with VPS18 antibody

This table summarizes the key differences between HOPS and CORVET complexes that can be leveraged for experimental discrimination of VPS18's complex-specific roles .

What emerging research questions about VPS18 remain to be addressed?

Despite significant advances in understanding VPS18 function, several critical questions remain at the forefront of research in this field. The mechanistic details of how VPS18 contributes to tethering complex assembly and membrane recognition remain incompletely understood, particularly regarding the structural determinants that enable selective recruitment to different endosomal compartments . The newly identified E3 ubiquitin ligase activity of VPS18 raises fundamental questions about its physiological substrates, regulatory mechanisms, and functional significance in the context of vesicular trafficking and beyond . The potential tissue-specific functions of VPS18 warrant deeper investigation, especially given its involvement in dendrite development of Purkinje cells, suggesting specialized roles in neuronal cells that may differ from its functions in other tissues . The precise contribution of VPS18 to human disease pathogenesis represents another critical frontier, as mutations or dysregulation of trafficking machinery components have been implicated in numerous disorders, yet the specific involvement of VPS18 in disease states remains largely unexplored .

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.