ATP6V1F Human

ATPase Transporting, Lysosomal V1 Subunit F Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function

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:

PropertyDetail
Gene LocationChromosome 7 (GRCh38.p14)
Protein DomainsV-ATPase F subunit family (Pfam: PF01992)
Subunit RoleEssential for V1 domain assembly and catalytic activity
Recombinant Form142 amino acids (including 23 His-tag residues), 15.8 kDa
Key InteractionsATP6V1D, ATP6V1B1/B2, ATP6V0A1, and ATP6V0D1/D2

The V-ATPase complex maintains organelle acidification (pH 4.5–6.0) required for:

  • Protein sorting and zymogen activation

  • Synaptic vesicle proton gradient generation

  • Lysosomal degradation and autophagy

Role in Hepatocellular Carcinoma (HCC)

ATP6V1F overexpression correlates strongly with HCC progression and prognosis:

Clinical Findings

Therapeutic ResponseATP6V1F-High HCCATP6V1F-Low HCC
Immune cell infiltration↓ CD8+ T cells↑ NK cells
ICI efficacy predictionPoor responseFavorable response

Experimental Interventions

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)

Disease Associations Beyond Cancer

  • 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

Research Tools and Reagents

Commercial recombinant ATP6V1F proteins enable mechanistic studies:

VendorCatalogPurityApplications
Prospec BioPRO-1466>95%Enzyme kinetics, Antibody production
Abcamab202196>90%SDS-PAGE, Mass spectrometry

Future Directions

Key unresolved questions include:

  • Structural basis of ATP6V1F-CANX interaction in calcium signaling

  • In vivo validation of DHA’s antitumor efficacy in HCC models

  • ATP6V1F’s role in immune checkpoint regulation (PD-1/PD-L1 axis)

Product Specs

Introduction
ATPase Transporting, Lysosomal V1 Subunit F (ATP6V1F), a crucial component of the vacuolar ATPase (V-ATPase), plays a vital role in the acidification of eukaryotic intracellular organelles. This multi-subunit enzyme is essential for various intracellular processes, including protein sorting, zymogen activation, receptor-mediated endocytosis, and the generation of synaptic vesicle proton gradients. V-ATPase comprises a cytosolic V1 domain and a transmembrane V0 domain. The V1 domain houses three A and three B subunits, along with two G subunits, and the C, D, E, F, and H subunits. Notably, the ATP catalytic site resides within the V1 domain. On the other hand, the V0 domain consists of five distinct subunits: a, c, c', c'', and d.
Description
Recombinant ATP6V1F Human, produced in E.Coli, is a single, non-glycosylated polypeptide chain comprising 142 amino acids (specifically, amino acids 1 to 119). With a molecular mass of 15.8 kDa, this protein features a 23 amino acid His-tag at its N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The ATP6V1F protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing Phosphate buffered saline (pH 7.4), 50% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product should be stored at 4°C. For extended storage, freezing at -20°C is recommended. To further enhance long-term stability, the addition of a carrier protein (either 0.1% HSA or BSA) is advisable. It is crucial to minimize freeze-thaw cycles to maintain product integrity.
Purity
The purity of the ATP6V1F protein is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
ATP6S14, VATF, Vma7, V-type proton ATPase subunit F, V-ATPase 14 kDa subunit.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAGRGKL IAVIGDEDTV TGFLLGGIGE LNKNRHPNFL VVEKDTTINE IEDTFRQFLN RDDIGIILIN QYIAEMVRHA LDAHQQSIPA VLEIPSKEHP YDAAKDSILR RARGMFTAED LR.

Q&A

What is ATP6V1F and what is its role in cellular physiology?

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

  • Generation of proton gradients in synaptic vesicles

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

How is ATP6V1F expression distributed across human tissues?

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 .

What experimental approaches are used to modulate ATP6V1F activity?

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)

How does ATP6V1F contribute to hepatocellular carcinoma pathogenesis?

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

  • Modulation of immune cell infiltration patterns

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

What is the relationship between ATP6V1F expression and immune checkpoint inhibitor therapy?

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

What signaling pathways mediate ATP6V1F's effects in cancer cells?

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

  • Cell migration and invasion 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

What are optimal controls when studying ATP6V1F in cellular models?

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

How can researchers differentiate direct ATP6V1F effects from general V-ATPase dysfunction?

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

What cellular models are most appropriate for ATP6V1F research?

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)

How should researchers interpret ATP6V1F expression data across different cancer types?

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:

    • Poorer OS and DFS in kidney renal papillary cell carcinoma (KIRP)

    • Better OS and DFS in brain lower grade glioma (LGG)

    • Worse OS and DFS in liver hepatocellular carcinoma (LIHC)

Analytical approaches should include:

  • Multi-cohort validation

  • Multivariate analyses controlling for confounding variables

  • Stratification by molecular subtypes

  • Integration with clinical parameters

What methods are recommended for integrating ATP6V1F data with broader V-ATPase complex function?

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

How can researchers address contradictory findings regarding ATP6V1F function?

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)

What are the challenges in detecting and quantifying ATP6V1F protein?

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

How should researchers design experiments to investigate ATP6V1F interaction with the immune system?

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 .

Product Science Overview

Introduction

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 .

Structure and Function

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 .

Genetic Information

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 .

Biological Significance

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 .

Clinical Relevance

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 .

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 2024 Thebiotek. All Rights Reserved.