GULP1 (GULP PTB Domain Containing Engulfment Adaptor 1) is a conserved adaptor protein critical for cellular processes such as apoptotic cell phagocytosis, lipid trafficking, and receptor-mediated endocytosis. It contains a phosphotyrosine-binding (PTB) domain that facilitates interactions with target proteins, including amyloid precursor protein (APP) and KEAP1, influencing pathways like amyloid-β production and NRF2 signaling .
Apoptotic Cell Clearance:
Lipid Trafficking:
Receptor Endocytosis:
GULP1 binds to the C-terminal domain of APP, enhancing its processing into amyloid-β (Aβ) peptides. Overexpression increases Aβ levels, while knockdown reduces them, suggesting a role in Alzheimer’s disease pathology .
| Experimental Model | Observation | Implication |
|---|---|---|
| APP–GAL4 Reporter | GULP1 enhances APP CTF and Aβ production | Potential therapeutic target for AD |
| Neuronal Co-localization | APP and GULP1 co-localize in neurons | In vivo relevance in neurodegeneration |
GULP1 binds KEAP1, stabilizing it in the cytoplasm and suppressing NRF2 nuclear translocation. In urothelial bladder cancer (UBC), GULP1 deficiency correlates with cisplatin resistance due to NRF2-driven antioxidant responses .
| Clinical Correlation | Mechanism | Outcome |
|---|---|---|
| GULP1 Promoter Hypermethylation | Silences GULP1 expression in UBC | Increased NRF2 activity, chemoresistance |
| Cisplatin Resistance | Observed in GULP1-low UBC cell lines/tumors | Poor patient prognosis |
Bladder Cancer:
| Expression Profile | Clinical Impact | Source |
|---|---|---|
| ERβ−/GULP1+ | Lower disease progression risk | |
| GULP1 Promoter Methylation | Detectable in urine (non-invasive biomarker) |
Alzheimer’s Disease:
Targeting GULP1: Inhibiting GULP1 may reduce Aβ production in Alzheimer’s, while restoring its expression could enhance chemosensitivity in UBC .
| Property | Specification | Use Case |
|---|---|---|
| Source | E. coli-derived, His-tagged | Structural studies, in vitro assays |
| Purity | >95% (SDS-PAGE) | Biochemical interactions |
| Stability | -20°C storage recommended | Long-term experiments |
| Antibody | Applications | Reactivity | Dilution |
|---|---|---|---|
| 19902-1-AP (Proteintech) | WB, IHC, ELISA | Human, Mouse | WB: 1:1000–1:8000 |
| IHC: 1:400–1:1600 |
GULP1 (engulfment adaptor protein 1) is the human counterpart of CED6 from Caenorhabditis elegans, with both playing a conserved role in cellular engulfment across species. GULP1 facilitates EphB/ephrinB trogocytosis—a process of cell surface material transfer—by collaborating with Tiam2 and is essential for recruiting dynamin, a crucial protein for cellular internalization . The protein contains a PTB (phosphotyrosine-binding) domain that mediates interactions with various cellular proteins, including the amyloid precursor protein (APP) .
GULP1 expression varies significantly between normal and pathological tissues. Analysis across different tissue types reveals that while GULP1 is generally downregulated in many cancer types, it shows specific and significant overexpression in hepatocellular carcinoma (HCC) . In HCC, GULP1 levels are markedly elevated compared to normal liver tissues, viral hepatitis tissues, nonalcoholic fatty liver disease tissues, and cirrhosis tissues, emphasizing its potential as a specific biomarker for distinguishing HCC from other liver conditions .
Several experimental approaches have proven effective for studying GULP1 expression and function:
Transcriptomic analysis: qRT-PCR for tissue and cell samples to quantify GULP1 mRNA levels
Protein detection: Western blotting and ELISA for protein quantification in tissues and serum
Spatial analysis: Confocal microscopy and high-throughput spatial transcriptomics to visualize GULP1 localization
Functional assays: Overexpression and knockdown studies to assess GULP1's impact on cellular processes
Interaction studies: Yeast two-hybrid systems to identify protein-protein interactions
GULP1 was identified as a novel APP C-terminal domain (APPc)-interacting protein through yeast two-hybrid screening. The interaction is specifically mediated by the NPTY motif of APP and the GULP1 PTB domain . Confocal microscopy studies have confirmed that a proportion of APP and GULP1 co-localize in neurons, supporting their functional interaction .
In experimental studies using an APP-GAL4 reporter assay, GULP1 significantly altered APP processing. Overexpression of GULP1 enhanced the generation of APP C-terminal fragments (CTFs) and amyloid-β peptide (Aβ), while knockdown of GULP1 suppressed both APP CTFs and Aβ production . This evidence suggests that GULP1 may play a significant role in Alzheimer's disease pathogenesis by influencing the production of Aβ, which is central to the development of the disease.
GULP1 demonstrates exceptional potential as both a diagnostic and predictive biomarker for HCC, offering several advantages over existing markers like alpha-fetoprotein (AFP) . The comparative diagnostic performance is summarized in the table below:
| Clinical Scenario | GULP1 (AUC) | AFP (AUC) | Statistical Significance |
|---|---|---|---|
| All HCC patients in high-risk group | 0.827 | 0.595 | P < 0.0001 |
| Early-stage HCC (mUICC I) | 0.749 | 0.516 | P < 0.05 |
| HCC vs. non-HCC liver tumors | 0.833 | 0.540 | P < 0.0001 |
| LC vs. non-HCC liver tumors | 0.790 | 0.524 | P = 0.002 |
GULP1 exhibits remarkably superior diagnostic accuracy compared to AFP, particularly in early-stage HCC detection, which is critical for improving patient outcomes . Additionally, GULP1 showed predictive accuracy for HCC recurrence comparable to that of a 15-gene risk score model, suggesting it can be used as a simpler and cost-effective biomarker without the need for complex multigene profiling .
Despite being typically recognized as a tumor suppressor in many cancers, GULP1 promotes tumor growth, epithelial-mesenchymal transition (EMT), and invasiveness in HCC by modulating β-catenin signaling . Research indicates that GULP1 likely influences these processes through ARF6 (ADP-ribosylation factor 6)-mediated β-catenin activation, though the exact molecular mechanisms require further characterization .
High-throughput spatial transcriptomics analyses have shown a pronounced increase in GULP1 levels specifically in malignant hepatocytes, suggesting a distinct spatial distribution pattern that contributes to its oncogenic role in HCC . This cancer-specific function highlights the context-dependent nature of GULP1's biological activities.
For researchers developing GULP1-based diagnostic tools, several methodological considerations are essential:
Current GULP1 research faces several challenges:
Fluctuating expression: GULP1 levels may vary due to liver inflammation, cirrhosis, and treatments, potentially affecting diagnostic reliability .
Mechanistic understanding: While GULP1's role in ARF6-mediated β-catenin activation has been identified, further targeted approaches are needed to definitively validate these interactions, particularly in disease contexts .
Translational barriers: Despite promising results as a biomarker, GULP1's modest predictive power in some contexts requires refinement of detection thresholds and validation methods before clinical implementation .
Context-dependent function: The contrasting roles of GULP1 across different cancer types (tumor suppressor in most cancers but oncogenic in HCC) necessitate tissue-specific research approaches .
Future GULP1 research should focus on:
Combinatorial biomarker panels: Exploring GULP1 in combination with AFP or other emerging markers to enhance predictive accuracy for cancer diagnosis and recurrence prediction .
Pathway interactions: Systematically investigating how GULP1 cooperates with Wnt, Notch, and Hedgehog pathways to influence disease progression .
Therapeutic development: Developing interventions targeting GULP1-driven ARF6–β-catenin signaling, particularly for controlling cancer progression and recurrence .
Cross-disease applications: Evaluating GULP1's potential role in other neurodegenerative or inflammatory conditions, given its established interaction with APP and role in cellular engulfment .
Non-invasive diagnostic refinement: Further developing liquid biopsy applications for GULP1 as a minimally invasive diagnostic tool with enhanced sensitivity and specificity .
When designing experiments to investigate GULP1:
Select appropriate model systems: Consider using both in vitro cell models and in vivo animal models that accurately represent the physiological context of interest (e.g., neurodegenerative disease models for APP interactions, hepatocellular models for cancer applications).
Employ complementary approaches: Combine genetic manipulation techniques (overexpression, knockdown) with functional assays to establish causative relationships between GULP1 and observed phenotypes .
Utilize spatial analysis: Incorporate confocal microscopy and spatial transcriptomics to understand GULP1's localization patterns, which may provide insights into its function in different cellular compartments .
Account for context dependency: Design experiments that acknowledge GULP1's contrasting roles across different tissues and disease states, with appropriate controls for each context .
The seemingly contradictory findings regarding GULP1's role as both a tumor suppressor in many cancers and an oncogene in HCC highlight the importance of context-dependent protein function . Researchers should:
Characterize tissue-specific interaction partners: Identify tissue-specific protein interactions that may explain differential functions.
Analyze post-translational modifications: Investigate whether different post-translational modifications of GULP1 exist across tissues.
Examine isoform expression: Determine if different isoforms of GULP1 are expressed in different tissues or disease states.
Consider microenvironment influences: Evaluate how the tissue microenvironment might influence GULP1 function through signaling pathway cross-talk.
Based on current evidence, the most promising clinical applications for GULP1 research include:
Early HCC detection: GULP1's superior performance in detecting early-stage HCC compared to AFP makes it a valuable biomarker for improving early diagnosis and patient outcomes .
Recurrence prediction: GULP1's ability to predict HCC recurrence could help guide treatment decisions and surveillance strategies for patients who have undergone initial treatment .
Therapeutic targeting: Developing strategies to modulate GULP1-driven pathways, particularly in HCC, could provide novel therapeutic approaches for controlling cancer progression .
Alzheimer's disease intervention: Given GULP1's role in APP processing and Aβ production, targeting this interaction could potentially influence Alzheimer's disease pathogenesis .
When conducting GULP1 research involving human subjects, researchers must adhere to standard ethical principles and regulatory requirements for human subjects research. These include:
Informed consent: Ensuring participants understand the research procedures, risks, benefits, and their rights before participating .
Privacy and confidentiality: Protecting participant data, potentially using Certificates of Confidentiality (CoC) for sensitive research .
Inclusion policies: Following NIH policies for inclusion of diverse populations across sexes/genders, races, ethnicities, and ages to ensure research benefits are broadly applicable .
IRB oversight: Obtaining appropriate Institutional Review Board approval, including consideration of single IRB requirements for multi-site research .
Risk minimization: Designing research to minimize risks to participants while maximizing potential benefits to participants and society .
The recombinant form of GULP1 is produced in Escherichia coli and is a single, non-glycosylated polypeptide chain containing 327 amino acids with a molecular mass of 36.9 kDa . It is fused to a 23 amino acid His-tag at the N-terminus and purified by proprietary chromatographic techniques .
GULP1 is involved in several biological processes, including:
GULP1’s role in these processes makes it a crucial protein for maintaining cellular homeostasis and efficient cellular function.