ATP6V1F encodes a 16 kDa protein (119 amino acids in its native form) that forms part of the peripheral V1 domain of V-ATPase . Key structural and functional attributes include:
The V-ATPase complex maintains organelle acidification (pH 4.5–6.0) required for:
ATP6V1F overexpression correlates strongly with HCC progression and prognosis:
Therapeutic Response | ATP6V1F-High HCC | ATP6V1F-Low HCC |
---|---|---|
Immune cell infiltration | ↓ CD8+ T cells | ↑ NK cells |
ICI efficacy prediction | Poor response | Favorable response |
Dihydroartemisinin (DHA), an antimalarial derivative, demonstrates inhibitory effects:
Reduces ATP6V1F expression by 58–72% in HuH-7 and Li-7 cells
Suppresses ATP production (↓ 2.1×) and NAD+/NADH ratio (↓ 1.8×)
Reverses CANX-mediated upregulation of ATP6V0B/ATP6V1F (p < 0.001)
Neurodegeneration: V-ATPase dysfunction caused by ATP6V0A1 mutations impairs lysosomal acidification, contributing to epileptic encephalopathies
Myopathies: Linked to limb-girdle muscular dystrophy type 2 and myofibrillar myopathy via calcium signaling defects
Commercial recombinant ATP6V1F proteins enable mechanistic studies:
Vendor | Catalog | Purity | Applications |
---|---|---|---|
Prospec Bio | PRO-1466 | >95% | Enzyme kinetics, Antibody production |
Abcam | ab202196 | >90% | SDS-PAGE, Mass spectrometry |
Key unresolved questions include:
ATP6V1F encodes a constituent of vacuolar ATPase (V-ATPase), a multi-subunit enzyme complex responsible for acidification of various cellular organelles. This proton pump is critical for maintaining pH homeostasis within organelles such as lysosomes, endosomes, and secretory vesicles. V-ATPase-mediated acidification is required for numerous essential cellular processes including:
Receptor-mediated endocytosis
Zymogen activation
Protein sorting and degradation
Methodologically, researchers investigating ATP6V1F function typically employ techniques including:
Gene silencing or overexpression to assess functional consequences
pH-sensitive fluorescent probes to measure organelle acidification
Co-immunoprecipitation to identify protein-protein interactions
Subcellular fractionation to determine localization patterns
ATP6V1F shows variable expression across tissues, with particularly high expression noted in metabolically active organs. Research approaches to characterize tissue-specific expression include:
Analysis of RNA-seq data from repositories like TCGA
Immunohistochemistry with validated antibodies
Western blotting of tissue lysates
Single-cell RNA sequencing to assess cell-type specific expression
Multiple database analyses (including TIMER, HCCDB, and GEPIA) have confirmed ATP6V1F expression across numerous tissue types, with significantly elevated levels observed in multiple cancer types compared to matched normal tissues .
Researchers employ several strategies to investigate ATP6V1F function:
Genetic approaches:
CRISPR/Cas9-mediated gene editing for knockout or knock-in models
siRNA or shRNA for transient or stable knockdown
Expression vectors for wild-type or mutant ATP6V1F overexpression
Pharmacological approaches:
V-ATPase inhibitors (e.g., bafilomycin A1, concanamycin)
pH-modulating compounds to assess compensatory mechanisms
Validation methods:
qRT-PCR and western blotting to confirm expression changes
Functional assays measuring organelle acidification
Phenotypic assessments (proliferation, migration, apoptosis)
ATP6V1F has been identified as overexpressed in hepatocellular carcinoma (HCC) with significant implications for disease progression. Research has revealed that:
Mechanistically, in vitro experiments have demonstrated that ATP6V1F promotes HCC progression through:
Enhanced cell proliferation and metastatic capacity
Inhibition of apoptotic pathways
Altered tumor microenvironment (TME) acidity
Research approaches to investigate these mechanisms include:
Wound healing and Transwell invasion assays
Apoptosis detection methods (flow cytometry, TUNEL assay)
Xenograft models with ATP6V1F modulation
Co-culture systems with immune cells
The potential role of ATP6V1F in immunotherapy response represents an emerging research area with clinical implications. Current evidence suggests:
ATP6V1F expression correlates with infiltration patterns of specific immune cell populations
High ATP6V1F expression is associated with altered expression of multiple immune checkpoints, including CTLA4, TIGIT, PDCD1, SIGLEC15, CD274, HAVCR2, LAG3, and PDCD1LG2
The Tumor Immune Dysfunction and Exclusion (TIDE) algorithm has been utilized to predict differential responses to immune checkpoint inhibitors based on ATP6V1F expression levels
Methodological approaches for investigating this relationship include:
Multiplex immunohistochemistry
Flow cytometry of tumor-infiltrating lymphocytes
Single-cell RNA sequencing of tumor microenvironment
Immune checkpoint blockade models with ATP6V1F manipulation
Research using Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and KEGG pathway analyses has identified several potential signaling mechanisms through which ATP6V1F may influence cellular behavior:
mTORC1 signaling pathway involvement
Autophagy regulation pathways
Lysosomal function pathways
Experimental approaches to investigate these pathways include:
Western blotting for pathway component activation
Pathway inhibitor studies
Proximity ligation assays for protein interactions
Phosphoproteomic analyses
Rigorous experimental design for ATP6V1F studies should include:
For genetic manipulation experiments:
Empty vector controls (for overexpression studies)
Non-targeting siRNA/shRNA (for knockdown studies)
Rescue experiments with wild-type ATP6V1F
Isogenic cell line pairs differing only in ATP6V1F status
For functional assays:
V-ATPase inhibitor controls
pH-matched controls when assessing acidification-dependent phenotypes
Time course experiments to detect temporal dynamics
Multiparameter assays to assess specificity of effects
Distinguishing ATP6V1F-specific effects from general V-ATPase dysfunction requires:
Comparative analysis with manipulation of other V-ATPase components
Structure-function studies with domain-specific mutations
Complementation assays with other V-ATPase subunits
Temporal control of ATP6V1F modulation (e.g., inducible systems)
Dose-response studies to identify threshold effects
Selection of experimental models should consider:
Cell line considerations:
HCC cell lines for cancer studies (e.g., HepG2, Huh7, MHCC97H)
Cell lines with varying baseline ATP6V1F expression
Primary cells to validate findings from established lines
3D culture systems to better recapitulate in vivo conditions
In vivo model considerations:
Tissue-specific conditional knockout models
Patient-derived xenografts
Genetically engineered mouse models with ATP6V1F alterations
Models that allow assessment of immune components (for TME studies)
ATP6V1F expression patterns vary across cancer types, requiring careful interpretation:
ATP6V1F is significantly overexpressed in multiple cancers, including BLCA, BRCA, CHOL, HNSC, ESCA, KICH, LIHC, KIRP, LUSC, READ, PRAD, SKCM, THCA, STAD, and UCEC
The prognostic significance of ATP6V1F differs by cancer type, with high expression predicting:
Analytical approaches should include:
Multi-cohort validation
Multivariate analyses controlling for confounding variables
Stratification by molecular subtypes
Integration with clinical parameters
Understanding ATP6V1F in the context of the complete V-ATPase complex requires:
Co-expression network analysis with other V-ATPase subunits
Protein-protein interaction studies
Comparative phenotypic analysis across V-ATPase subunit alterations
Systems biology approaches integrating transcriptomic, proteomic, and functional data
Research has shown that different V-ATPase subunits can be associated with distinct clinical entities:
ATP6V0A1 mutations cause developmental and epileptic encephalopathy
ATP6V1C1 and ATP6V1B2 variants are linked to DOORS syndrome and related disorders
When encountering seemingly contradictory results across studies, consider:
Tissue-specific or context-dependent effects
Differences in experimental models or conditions
Technical variations in ATP6V1F detection or modulation
Threshold effects in ATP6V1F expression levels
Compensatory mechanisms within the V-ATPase complex
Methodological approaches to resolve contradictions include:
Systematic replication studies
Meta-analysis of published data
Direct comparison of experimental conditions
Exploration of biological variables (cell type, genetic background)
Accurate ATP6V1F protein analysis requires attention to:
Antibody validation (using knockout controls)
Appropriate subcellular fractionation techniques
Consideration of post-translational modifications
Comparative analysis of protein vs. mRNA levels
Recommended approaches include:
Multiple antibody validation
Inclusion of recombinant protein standards
Mass spectrometry-based quantification
Assessment of protein complex assembly
Investigating the immunomodulatory effects of ATP6V1F requires:
Co-culture systems with relevant immune cell populations
Flow cytometry to characterize immune infiltrates
Cytokine profiling before and after ATP6V1F modulation
In vivo models with intact immune systems
Data from HCC studies suggest significant correlations between ATP6V1F expression and immune checkpoint molecules, indicating complex interactions with the immune microenvironment that warrant careful experimental design .
ATPase Transporting, Lysosomal V1 Subunit F, also known as ATP6V1F, is a crucial component of the vacuolar ATPase (V-ATPase) complex. This enzyme complex is responsible for the acidification of intracellular organelles in eukaryotic cells, which is essential for various cellular processes such as protein sorting, zymogen activation, receptor-mediated endocytosis, and synaptic vesicle proton gradient generation .
The V-ATPase complex is composed of two main domains: the cytosolic V1 domain and the transmembrane V0 domain. The V1 domain, where ATP6V1F is located, consists of multiple subunits including three A and three B subunits, two G subunits, and the C, D, E, F, and H subunits. The V1 domain is responsible for ATP hydrolysis, which provides the energy required for proton translocation .
ATP6V1F specifically is a 14 kDa subunit within the V1 domain. It plays a critical role in the assembly and stability of the V-ATPase complex. The V0 domain, on the other hand, is responsible for proton translocation across the membrane and consists of five different subunits: a, c, c’, c", and d .
The ATP6V1F gene is located on chromosome 7q32.1 and encodes a protein that is predominantly hydrophilic. The gene has been identified through large-scale sequencing of a human fetal brain cDNA library. The encoded protein shares significant sequence identity with homologs in other species, such as Manduca sexta and Drosophila .
The acidification of intracellular organelles mediated by V-ATPase is vital for numerous cellular functions. For instance, it is involved in the degradation of macromolecules within lysosomes, the maturation of endosomes, and the generation of synaptic vesicle proton gradients necessary for neurotransmitter loading. Dysregulation of V-ATPase activity can lead to various diseases, including neurodegenerative disorders and cancer .
Mutations in the ATP6V1F gene have been associated with several genetic disorders. Notably, it has been linked to Spastic Paraplegia 2, X-Linked, and Myotonia Congenita, Autosomal Dominant. These conditions highlight the importance of ATP6V1F in maintaining normal cellular function and the potential consequences of its dysfunction .