Recombinant Rat V-type proton ATPase subunit e 1 (Atp6v0e1)

Shipped with Ice Packs
In Stock

Description

Expression Systems and Product Variants

Product Code (CUSABIO)SourceTagPurityApplications
CSB-CF755280RAE. coliNone≥85% WB, ELISA, Functional Assays
CSB-EP755280RA1Mammalian cellsHis-tag>90% Structural Studies

The protein is typically lyophilized in Tris/PBS-based buffer (pH 8.0) with trehalose for stability .

Research Applications

Recombinant Atp6v0e1 is widely used to study V-ATPase mechanisms and associated pathologies:

  • Functional Studies:

    • Investigating proton transport defects in lysosomal storage disorders .

    • Characterizing pH-dependent autophagy regulation .

  • Disease Modeling:
    Mutations in V-ATPase subunits (e.g., ATP6V0A1) are linked to neurological disorders like developmental epileptic encephalopathy (DEE). Recombinant Atp6v0e1 aids in dissecting these mechanisms .

  • Structural Biology:
    Used in cryo-EM studies to resolve V<sub>0</sub> domain architecture and subunit interactions .

Key Findings from Recent Studies

  • Role in Neurodegeneration:
    Impaired V-ATPase function due to subunit mutations disrupts lysosomal acidification, leading to autophagosome accumulation and neurodegeneration .

  • Interaction Networks:
    Atp6v0e1 cooperates with subunits a1 and d to stabilize the V<sub>0</sub> complex, as shown in crosslinking assays .

Future Directions

Ongoing research focuses on:

  • Engineering thermostable mutants for crystallography.

  • Developing inhibitors targeting V<sub>0</sub> subunits for cancer therapy .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, we can fulfill specific format requests. Please indicate your desired format in the order notes, and we will accommodate your needs.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Note: All our proteins are shipped with standard blue ice packs. For dry ice shipping, please inform us in advance, as additional fees will apply.
Notes
Repeated freezing and thawing is discouraged. Store working aliquots at 4°C for up to one week.
Reconstitution
Prior to opening, briefly centrifuge the vial to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration between 0.1 and 1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol final concentration is 50%. Customers may use this as a reference.
Shelf Life
The shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. Lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C, and aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you have a specific tag type requirement, please inform us, and we will prioritize developing the specified tag.
Synonyms
Atp6v0e1; Atp6v0e; Dsr1; V-type proton ATPase subunit e 1; V-ATPase subunit e 1; D-serine-regulated transcript 1 protein; DSR-1; V-ATPase 9.2 kDa membrane accessory protein; V-ATPase M9.2 subunit; Vacuolar proton pump subunit e 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-81
Protein Length
Full Length of Mature Protein
Species
Rattus norvegicus (Rat)
Target Names
Target Protein Sequence
AYHGLTVPLIVMSVFWGFVGLLVPWFIPKGPNRGVIITMLVTCSVCCYLFWLIAILAQLN PLFGPQLKNETIWYLKYHWP
Uniprot No.

Target Background

Function
Vacuolar ATPase plays a crucial role in acidifying various intracellular compartments in eukaryotic cells.
Database Links

KEGG: rno:94170

STRING: 10116.ENSRNOP00000004653

UniGene: Rn.789

Protein Families
V-ATPase e1/e2 subunit family
Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What is the structure and function of V-type proton ATPase subunit e 1 in rats?

V-type proton ATPase (V-ATPase) is a multisubunit enzyme complex composed of two major structural domains: a peripheral V1 complex that hydrolyzes ATP and a membrane-integrated V0 complex that translocates protons across membranes. The V1 ATPase contains three copies each of subunits A, B, E, and G, plus single copies of subunits C, D, F, and H, which together form the catalytic machinery for ATP hydrolysis . In contrast, the V0 proton pump, where subunit e1 is located, comprises subunits a, c8, c', c'', d, e, f, and V0a1, which collectively form the proton translocation pathway across membranes .

The subunit e1 (Atp6v0e1) represents one of the smaller components of the V0 domain and contributes to the structural integrity of the proton channel. Although smaller than many other V-ATPase components, this subunit plays critical roles in the assembly and stability of the entire complex. In rat models, this protein functions primarily in acidifying intracellular compartments such as lysosomes, endosomes, and secretory vesicles, and in some specialized cells, it may also participate in plasma membrane-associated proton transport.

The primary function of V-ATPase complexes containing Atp6v0e1 involves maintaining organelle pH homeostasis, which is essential for numerous cellular processes including protein sorting, receptor-mediated endocytosis, neurotransmitter uptake, and lysosomal degradation. In specialized tissues, these complexes may also participate in processes such as bone resorption, renal acid secretion, and pH regulation in the brain microenvironment.

How does rat Atp6v0e1 compare to human ATP6V0E1 in terms of structure and function?

Rat Atp6v0e1 and human ATP6V0E1 share significant sequence homology, reflecting their evolutionarily conserved functions across mammalian species. Both proteins serve as components of the V0 domain within the V-ATPase complex, contributing to proton translocation mechanisms. The high degree of conservation suggests fundamental importance in cellular physiology across species.

Structurally, both proteins are relatively small subunits integrated within the membrane-embedded V0 complex. They share similar hydrophobic domains that facilitate membrane anchoring and interactions with adjacent subunits within the complex. Functional studies indicate that both rat and human variants participate in maintaining the structural integrity of the V-ATPase complex and supporting its proton pumping activities.

When designing experiments to express recombinant rat Atp6v0e1, understanding these cross-species similarities and differences becomes crucial for properly interpreting results and developing appropriate experimental controls. This is especially relevant when using heterologous expression systems or when developing targeting strategies for research applications.

What expression systems are most effective for producing recombinant rat Atp6v0e1?

Several expression systems can be utilized for generating recombinant rat Atp6v0e1, each with distinct advantages depending on research objectives. For mammalian expression, recombinant adeno-associated virus (rAAV) vectors offer significant benefits, particularly for in vivo applications. Among various serotypes, rAAV6 has demonstrated impressive transduction efficiency in rat cells, making it potentially valuable for Atp6v0e1 expression studies .

When using viral vectors for Atp6v0e1 expression, promoter selection becomes critical. Studies with similar membrane proteins have utilized CMV promoters for strong expression, though tissue-specific promoters may provide more targeted expression when needed . For rAAV-based delivery systems, packaging capabilities typically accommodate the relatively small Atp6v0e1 coding sequence, allowing for efficient viral production with titers reaching 10^5 genome copies per cell for effective transduction .

Bacterial expression systems, while cost-effective, present challenges for membrane proteins like Atp6v0e1 due to potential folding issues and lack of post-translational modifications. If pursuing bacterial expression, specialized strains and fusion partners designed for membrane proteins should be considered, though mammalian expression systems generally provide superior results for functional studies of V-ATPase components.

What are the optimal viral vector systems for in vivo delivery of rat Atp6v0e1 to specific cell types?

For targeted delivery of rat Atp6v0e1 to specific cell populations in vivo, selection of the appropriate viral vector system is crucial. Recombinant adeno-associated virus (rAAV) represents one of the most promising platforms due to its safety profile, low immunogenicity, and ability to transduce both dividing and non-dividing cells . Among the various rAAV serotypes, serotype-specific tropism significantly influences transduction efficiency in target tissues.

Research examining rAAV serotypes 1, 2, 5, 6, 7, 8, and 9 has demonstrated distinct cellular preferences, with rAAV6 showing particularly strong tropism for rat astrocytes (>90% infection rate in virus-infected areas) while exhibiting lower neuronal infection rates (~10%) . In contrast, rAAV2 preferentially transduces neurons (~65%) with lower astrocytic infection rates (~20%) . This differential tropism becomes especially important when designing experiments targeting specific cell populations for Atp6v0e1 expression or knockdown.

When targeting non-neural tissues, serotype selection should be tailored accordingly. For example, rAAV8 and rAAV9 demonstrate strong liver tropism, while rAAV1 and rAAV7 may be preferred for skeletal muscle. Importantly, the packaging capacity of all rAAV serotypes (~4.7 kb) comfortably accommodates the Atp6v0e1 coding sequence, allowing inclusion of regulatory elements and reporter genes within a single construct.

Beyond serotype selection, combining viral vectors with cell-type-specific promoters substantially enhances targeting precision. For astrocyte-specific expression of Atp6v0e1, pairing rAAV6 with GFAP promoter elements would maximize specificity, while neuronal targeting might employ rAAV2 with synapsin or CaMKII promoters. Additionally, incorporating miRNA target sequences that suppress expression in non-target cells can further refine specificity profiles.

How can researchers effectively measure functional activity of recombinant rat Atp6v0e1 in experimental systems?

Assessing the functional activity of recombinant rat Atp6v0e1 requires methodologies that evaluate both its incorporation into V-ATPase complexes and the resulting proton pumping activity. Since Atp6v0e1 functions as part of a multisubunit complex, functional assays must extend beyond simple protein expression confirmation to assess physiological activity within cellular contexts.

One robust approach involves using pH-sensitive fluorescent probes to measure acidification of intracellular compartments. Probes such as LysoTracker, LysoSensor, or pHrodo can be employed to visualize and quantify changes in organelle pH following expression of recombinant Atp6v0e1. These measurements can be performed in live cells using confocal microscopy or flow cytometry, providing real-time assessment of V-ATPase function. Complementary techniques include pH measurements using ratiometric probes (such as BCECF) targeted to specific cellular compartments through fusion with organelle-targeting sequences.

For biochemical verification of proper complex assembly, co-immunoprecipitation assays using antibodies against other V-ATPase subunits can confirm whether recombinant Atp6v0e1 physically associates with its native partner proteins. This should be complemented with subcellular fractionation and western blotting to verify correct membrane localization of the recombinant protein. When antibodies against rat Atp6v0e1 are unavailable, epitope tagging (such as HA or FLAG) can facilitate detection, though careful validation is necessary to ensure tags don't interfere with assembly or function.

ATP hydrolysis assays using isolated membrane fractions represent another valuable approach for functional assessment. By comparing ATPase activity in the presence and absence of specific V-ATPase inhibitors (such as bafilomycin A1 or concanamycin A), researchers can quantify the contribution of fully assembled V-ATPase complexes containing the recombinant Atp6v0e1. This approach can be particularly informative when coupled with genetic knockdown/knockout strategies to demonstrate functional replacement by the recombinant protein.

What strategies can overcome challenges in expressing and purifying functional rat Atp6v0e1?

Expressing and purifying functional rat Atp6v0e1 presents several challenges inherent to membrane proteins. As a component of a multisubunit complex, Atp6v0e1 may exhibit limited stability and functionality when expressed in isolation. Several strategies can address these challenges to improve experimental outcomes.

Co-expression with partner subunits represents one of the most effective approaches for obtaining properly folded and functional Atp6v0e1. By simultaneously expressing multiple V-ATPase subunits, particularly those directly interacting with subunit e1 within the V0 domain, researchers can facilitate proper complex assembly. This approach often requires multi-cistronic expression constructs or co-transfection of multiple plasmids. When using viral vectors such as rAAV, careful consideration of packaging limits becomes necessary, potentially requiring the use of multiple complementary vectors administered simultaneously.

Fusion with stability-enhancing partners can significantly improve expression and purification outcomes. Strategic placement of tags (such as His, GST, or MBP) can enhance protein solubility and provide affinity purification handles without compromising function. For structure-function studies requiring removal of these tags, inclusion of specific protease recognition sequences enables tag removal under controlled conditions. Additionally, incorporating fluorescent proteins (such as GFP) allows real-time monitoring of expression and trafficking when placed at termini less critical for function.

When expressing Atp6v0e1 in heterologous systems, careful selection of detergents for membrane protein extraction is crucial. Mild non-ionic detergents (such as DDM, LMNG, or digitonin) generally prove more successful in maintaining native protein conformations compared to harsher ionic detergents. Detergent screening panels should be empirically tested to identify optimal conditions that balance extraction efficiency with protein stability.

For functional reconstitution, incorporation into artificial membrane systems (such as proteoliposomes, nanodiscs, or polymeric nanomembranes) can provide near-native environments for biochemical and biophysical characterization. These systems allow precise control over lipid composition, which may significantly impact V-ATPase assembly and activity. Such reconstitution approaches become particularly valuable when coupled with functional assays measuring proton translocation across these artificial membranes.

How can recombinant rat Atp6v0e1 be used for studying V-ATPase assembly and regulation?

Recombinant rat Atp6v0e1 provides a powerful tool for investigating V-ATPase assembly dynamics and regulatory mechanisms. By incorporating modifications such as fluorescent tags or specific mutations into the recombinant protein, researchers can track assembly pathways, interaction partners, and regulatory events in real-time and under various physiological conditions.

Fluorescence resonance energy transfer (FRET) approaches utilizing differentially labeled V-ATPase subunits (including tagged Atp6v0e1) can reveal spatial relationships and conformational changes during complex assembly and activation. This technique is particularly valuable for monitoring dynamic interactions between Atp6v0e1 and other V0 domain components under different cellular conditions, such as nutrient availability, stress, or pharmacological interventions. When combined with live-cell imaging, these approaches provide unprecedented temporal resolution of assembly and disassembly events.

Site-directed mutagenesis of recombinant Atp6v0e1 allows systematic investigation of structure-function relationships. By introducing specific mutations in potential interaction interfaces or regulatory motifs, researchers can identify residues critical for incorporation into the V-ATPase complex or for responding to cellular signaling events. Such mutational analysis becomes particularly informative when combined with cryo-electron microscopy of partially assembled complexes, enabling correlation between structural features and functional outcomes.

Proximity labeling approaches, such as BioID or APEX2 fused to recombinant Atp6v0e1, facilitate identification of transient interaction partners that may regulate V-ATPase assembly or activity. These techniques enable labeling of proteins within nanometer-scale proximity of Atp6v0e1 in living cells, followed by purification and mass spectrometry identification of the labeled proteins. This approach can reveal novel regulatory interactions that might be missed by conventional co-immunoprecipitation methods due to their transient or weak nature.

Domain swapping experiments between rat Atp6v0e1 and homologs from other species or between different isoforms can identify regions responsible for specific functional properties or regulatory responses. By creating chimeric proteins and assessing their incorporation into V-ATPase complexes and subsequent functional outcomes, researchers can map domains critical for proper assembly, membrane targeting, and response to regulatory signals.

What experimental designs best evaluate tissue-specific functions of rat Atp6v0e1?

Investigating tissue-specific functions of rat Atp6v0e1 requires experimental designs that address both expression patterns and functional consequences in diverse cellular contexts. Combined approaches utilizing recombinant expression systems, genetic manipulation, and physiological measurements provide comprehensive insights into tissue-specific roles.

Targeted delivery of recombinant Atp6v0e1 to specific tissues using viral vectors with appropriate tropism represents a powerful approach. The selection of rAAV serotypes based on their differential tropism can significantly enhance targeting precision, with rAAV6 showing particularly strong astrocyte tropism in the rat brain, while other serotypes preferentially target neurons or peripheral tissues . Coupling these vectors with tissue-specific promoters further enhances specificity, allowing precise manipulation of Atp6v0e1 levels in targeted cell populations.

For loss-of-function studies, RNA interference or CRISPR-Cas9 approaches enable tissue-specific knockdown or knockout of endogenous Atp6v0e1. When delivered through viral vectors like rAAV6, shRNA constructs targeting Atp6v0e1 can achieve efficient gene silencing in specific cell types such as astrocytes . These approaches can be particularly valuable when combined with rescue experiments using recombinant Atp6v0e1 variants resistant to the silencing mechanism, allowing determination of structure-function relationships in native cellular contexts.

Physiological measurements tailored to specific tissues provide functional readouts of V-ATPase activity. In neurons, electrophysiological recordings can assess synaptic vesicle acidification and neurotransmitter loading. In renal tissues, measurements of urinary pH and ammonium excretion can evaluate proton secretion functions. For osteoclasts, bone resorption assays provide functional assessment of V-ATPase-dependent activities. These tissue-specific functional assays should be coupled with subcellular localization studies of recombinant Atp6v0e1 to correlate protein distribution with functional outcomes.

Ex vivo tissue preparations and organoid models offer intermediate complexity between cell culture and in vivo systems. Primary astrocyte cultures, which can be efficiently transduced with rAAV6 vectors , provide an excellent platform for investigating Atp6v0e1 functions in this cell type. Similarly, hippocampal slice cultures or renal tubule preparations maintain tissue architecture while allowing experimental manipulation and detailed functional measurements that might be challenging in intact animals.

How do post-translational modifications affect rat Atp6v0e1 function and stability?

Post-translational modifications (PTMs) of rat Atp6v0e1 represent critical regulatory mechanisms affecting protein stability, subcellular trafficking, complex assembly, and functional activity. Understanding these modifications provides insights into V-ATPase regulation and potential therapeutic targets for conditions involving aberrant V-ATPase function.

Phosphorylation represents one of the most prevalent regulatory PTMs affecting V-ATPase subunits. Phosphoproteomic analyses have identified several potential phosphorylation sites on V-ATPase components, including subunit e isoforms. These modifications can influence subunit interactions, conformational states, or association with regulatory proteins. To investigate phosphorylation effects on recombinant Atp6v0e1, researchers can employ phosphomimetic mutations (such as serine/threonine to aspartate substitutions) or phosphodeficient mutations (serine/threonine to alanine substitutions) combined with functional assays to determine how specific phosphorylation events impact activity.

Ubiquitination and SUMOylation of V-ATPase components regulate complex stability and turnover. By creating recombinant Atp6v0e1 variants with mutations at potential ubiquitination sites, researchers can assess how these modifications influence protein half-life, complex assembly, and subcellular distribution. These studies can be complemented with proteasome inhibitors or deubiquitinating enzyme modulators to further characterize the role of the ubiquitin-proteasome system in regulating Atp6v0e1 levels and activity.

Glycosylation may affect Atp6v0e1 folding, trafficking, and stability. Although not all V-ATPase subunits undergo glycosylation, potential N-linked glycosylation sites can be investigated through site-directed mutagenesis of asparagine residues within consensus sequences. Comparing wild-type Atp6v0e1 with glycosylation-deficient variants in terms of trafficking efficiency, complex incorporation, and functional activity provides insights into the importance of this modification. Additionally, treating cells with glycosylation inhibitors (such as tunicamycin) or specific glycosidases can reveal how glycan structures contribute to protein function.

Protein-protein interaction networks often depend on PTMs that create or mask binding interfaces. Using recombinant Atp6v0e1 with mutations at key modification sites, coupled with interactome analysis techniques such as co-immunoprecipitation followed by mass spectrometry, researchers can map how specific PTMs influence the assembly of V-ATPase complexes and association with regulatory proteins. These studies become particularly informative when conducted under different physiological conditions that trigger known regulatory responses, such as glucose starvation or hormonal stimulation.

How should researchers analyze data from studies comparing wild-type and mutant forms of rat Atp6v0e1?

Expression level normalization represents a critical first step in comparative analysis. Western blotting with quantitative densitometry or flow cytometry for tagged variants can establish relative expression levels between wild-type and mutant forms. This normalization becomes essential for distinguishing between phenotypes resulting from functional deficits versus those stemming from expression differences. When possible, creating stable cell lines with inducible expression systems helps minimize variation in expression levels and timing, reducing experimental noise.

For functional comparisons, multiple independent parameters should be assessed, including: (1) protein stability and half-life, (2) subcellular localization and trafficking, (3) incorporation into V-ATPase complexes, (4) enzymatic activity (ATP hydrolysis), and (5) proton pumping efficiency. Each parameter provides distinct insights, and discrepancies between these measurements may reveal specific mechanistic defects. For example, mutations affecting complex assembly would impact both ATP hydrolysis and proton pumping, while mutations specifically disrupting the proton pathway might affect pumping without altering ATP hydrolysis.

Statistical analysis should account for biological variability inherent in multisubunit membrane protein complexes. Multiple biological replicates (typically n≥5) are necessary, with data presented as mean ± standard error of the mean or with appropriate nonparametric representations if data do not meet normality assumptions. Two-way ANOVA with appropriate post-hoc tests often proves valuable for comparing multiple mutants across different functional parameters, enabling identification of statistically significant differences while controlling for multiple comparisons.

Structure-function correlations require integration of mutational data with available structural information. When specific mutations cause functional defects, these should be mapped onto structural models of the V-ATPase complex to identify potential mechanisms. Homology modeling based on published V-ATPase structures can provide valuable insights even when rat-specific structures are unavailable. Additionally, evolutionary conservation analysis of affected residues across species can further support the functional importance of specific domains or motifs.

What controls and validations are essential when using antibodies against rat Atp6v0e1?

When utilizing antibodies against rat Atp6v0e1 for research applications, comprehensive validation and proper controls are essential to ensure specificity, sensitivity, and reproducibility. Given the challenges associated with generating antibodies against small membrane protein subunits, rigorous validation becomes particularly critical for reliable experimental outcomes.

Specificity validation should employ multiple complementary approaches. Western blotting with samples from tissues expressing varying levels of Atp6v0e1 can establish signal correlation with expected expression patterns. Critically, specificity should be confirmed using negative controls where Atp6v0e1 is absent or depleted, such as through CRISPR knockout or siRNA knockdown. Additionally, signal detection in heterologous expression systems (such as HEK293 cells) transfected with rat Atp6v0e1 provides further validation. For polyclonal antibodies, pre-adsorption with the immunizing peptide should eliminate specific signal, providing another specificity control.

Cross-reactivity assessment against related proteins is essential, particularly with other V-ATPase subunits or isoforms that may share sequence homology. This evaluation becomes especially important when studying specific isoforms in tissues expressing multiple variants. Comparative immunoblotting or immunostaining using samples with differential expression of related proteins helps establish antibody specificity among closely related targets.

Application-specific validation ensures antibody performance in specific experimental contexts. For immunohistochemistry or immunofluorescence applications, antibodies should be tested with appropriate fixation protocols, as some epitopes may be sensitive to particular fixatives or antigen retrieval methods. Similarly, for immunoprecipitation applications, validation should confirm the ability to capture native Atp6v0e1 from lysates under conditions that preserve relevant protein-protein interactions.

When commercial antibodies are employed, researchers should critically evaluate provided validation data while performing independent validation in their specific experimental systems. The quality of commercially available antibodies against V-ATPase subunits can vary significantly, and published literature should be consulted for previously validated reagents. For example, antibody resources like Antibodypedia or the Antibody Registry can provide community feedback on specific antibodies for rat V-ATPase components.

Batch-to-batch variation monitoring becomes crucial for long-term studies, particularly with polyclonal antibodies. New antibody lots should be validated against previous lots using identical samples and protocols to ensure consistent performance. For critical experiments, researchers should consider reserving sufficient antibody from a single validated lot, especially for longitudinal studies where consistent detection is essential for comparative analyses.

How can researchers troubleshoot issues with recombinant rat Atp6v0e1 expression and function?

Troubleshooting recombinant rat Atp6v0e1 expression and functional issues requires systematic approaches addressing the various challenges associated with membrane protein biology. By methodically evaluating each stage from gene delivery to functional assessment, researchers can identify and resolve specific bottlenecks in their experimental systems.

For viral vector-mediated expression, infection efficiency represents a common challenge. When using rAAV vectors, researchers should verify viral titers through qPCR and optimize multiplicity of infection (MOI) through dose-response experiments . Different cell types may require varying MOIs for optimal expression without toxicity. Additionally, serotype selection significantly impacts transduction efficiency, with rAAV6 demonstrating superior performance in certain cell types such as astrocytes . If expression levels remain suboptimal despite high MOIs, alternative promoters or enhancer elements may improve transcriptional activity.

Protein misfolding or aggregation often occurs with membrane proteins expressed outside their native membrane environment. To address this, researchers can modify culture conditions by lowering incubation temperatures (28-30°C instead of 37°C) to slow folding and reduce aggregation. Additionally, incorporating chemical chaperones such as glycerol, DMSO, or 4-phenylbutyrate into culture media may enhance proper folding. Co-expression with interacting V-ATPase subunits can also promote proper folding by providing stabilizing protein-protein interactions that mimic the native complex environment.

Membrane localization issues may manifest as cytoplasmic accumulation of recombinant Atp6v0e1. This could result from saturation of cellular trafficking machinery or absence of necessary targeting signals. Subcellular fractionation followed by western blotting can identify where protein accumulation occurs. If proteins accumulate in the ER, as indicated by co-localization with ER markers, modifications such as optimizing signal sequences or co-expressing trafficking chaperones may improve membrane integration.

Functional deficits despite successful expression and localization often indicate problems with V-ATPase complex assembly or activation. Researchers should verify association with other V-ATPase subunits through co-immunoprecipitation experiments. If recombinant Atp6v0e1 fails to incorporate into complexes, examining post-translational modifications or potential steric hindrance from affinity tags may provide insights. Additionally, ensuring proper assembly of the entire V-ATPase complex requires presence of all essential subunits, which may necessitate supplementation or co-expression strategies in certain experimental systems.

When applying rAAV-mediated expression in vivo, brain region-specific or cell type-specific expression patterns may require optimization. Stereotaxic injection parameters including volume, rate, and coordinates significantly impact viral spread and expression patterns . For targeting specific cell populations, combining appropriate rAAV serotypes with cell-type-specific promoters enhances specificity. For example, rAAV6 with astrocyte-specific promoters provides excellent astrocyte targeting, while other combinations may better suit neuronal expression .

What emerging technologies will advance research on rat Atp6v0e1 function in disease models?

Emerging technologies across molecular biology, imaging, and computational domains promise to revolutionize research on rat Atp6v0e1 function in disease models. These advanced approaches will enable more precise manipulation and visualization of V-ATPase complexes in physiologically relevant contexts, leading to deeper mechanistic insights and potential therapeutic applications.

CRISPR-based technologies beyond basic gene editing offer unprecedented precision for studying Atp6v0e1. Base editors and prime editors allow introduction of specific mutations without double-strand breaks, enabling subtle modifications that preserve reading frames and avoid compensatory mechanisms often triggered by complete knockouts. CRISPRa (activation) and CRISPRi (interference) systems permit temporal control over Atp6v0e1 expression without permanent genetic modifications, allowing researchers to assess acute versus chronic effects of expression changes. When delivered via appropriately targeted viral vectors such as rAAV6, these tools can achieve cell-type-specific manipulations in complex tissues .

Advanced imaging techniques with enhanced spatiotemporal resolution will transform our ability to visualize V-ATPase dynamics. Super-resolution microscopy approaches such as STORM, PALM, or STED break the diffraction limit, enabling visualization of individual V-ATPase complexes within specific membrane microdomains. When combined with split-fluorescent protein technologies, these approaches can visualize Atp6v0e1 incorporation into assembling V-ATPase complexes in real-time. Additionally, expansion microscopy physically enlarges samples while maintaining molecular relationships, potentially revealing previously unresolvable details of complex architecture.

Organoid and microphysiological systems (MPS) provide physiologically relevant contexts for studying Atp6v0e1 functions in disease models. Brain organoids derived from rat neural stem cells, potentially transduced with rAAV vectors expressing or silencing Atp6v0e1, can recapitulate developmental processes and circuit formation in three-dimensional contexts. Similarly, kidney or bone organoids can model tissue-specific functions of V-ATPase in pH regulation or bone remodeling. MPS approaches integrating multiple organoids with controlled fluid flow enable assessment of systemic consequences of V-ATPase dysfunction across interconnected tissues.

Computational approaches including molecular dynamics simulations and systems biology models will increasingly contribute to understanding Atp6v0e1 functions. As structural data on V-ATPase complexes expands, atomic-level simulations can predict how specific mutations or post-translational modifications affect proton translocation mechanisms or subunit interactions. Network analysis integrating transcriptomic, proteomic, and metabolomic data can identify broader consequences of V-ATPase dysfunction in disease states, revealing compensatory mechanisms or identifying potential therapeutic intervention points.

Single-cell multi-omics approaches will reveal cell-type-specific functions and responses related to Atp6v0e1. By combining single-cell transcriptomics, proteomics, and metabolomics from tissues expressing manipulated Atp6v0e1 levels, researchers can identify differential responses across heterogeneous cell populations. These approaches become particularly powerful when applied to brain tissues where diverse neuronal and glial populations may exhibit distinct dependencies on V-ATPase functions, potentially explaining selective vulnerability in neurodegenerative conditions.

How might understanding rat Atp6v0e1 contribute to therapeutic development for V-ATPase-related disorders?

Understanding rat Atp6v0e1 biology holds significant potential for therapeutic development targeting V-ATPase-related disorders. As research uncovers the structural determinants and regulatory mechanisms of this subunit, opportunities emerge for designing targeted interventions with improved specificity and reduced side effects compared to current approaches.

Structure-guided drug design targeting specific V-ATPase subunit interactions represents a promising therapeutic strategy. As high-resolution structures of V-ATPase complexes become available, computational approaches can identify potential binding pockets at subunit interfaces, including those involving Atp6v0e1. Small molecules designed to stabilize or disrupt specific interactions could modulate V-ATPase assembly or activity with greater selectivity than current inhibitors that target the catalytic machinery. These approaches could be particularly valuable for conditions where tissue-specific V-ATPase modulation is desired, such as osteoporosis (targeting osteoclasts) or certain renal tubular acidosis variants.

Gene therapy approaches utilizing rAAV vectors show considerable promise for addressing genetic V-ATPase deficiencies. The demonstrated efficacy of rAAV6 for astrocyte-targeted gene delivery in rat models provides a foundation for developing therapies targeting CNS manifestations of V-ATPase disorders . For conditions involving loss-of-function mutations in Atp6v0e1, rAAV-mediated gene replacement could restore functional protein expression in affected tissues. Conversely, for conditions where excessive V-ATPase activity contributes to pathology, RNA interference delivered via appropriate viral vectors could achieve targeted knockdown.

Post-translational modification pathways regulating Atp6v0e1 offer additional therapeutic targets. By identifying kinases, phosphatases, ubiquitin ligases, or deubiquitinating enzymes that modulate Atp6v0e1 stability or function, researchers can develop inhibitors targeting these regulatory enzymes. Such approaches could tune V-ATPase activity without directly targeting the complex itself, potentially offering more physiological modulation. This strategy becomes particularly relevant for conditions involving dysregulated (rather than absent) V-ATPase function.

Targeted protein degradation technologies, including PROTACs (Proteolysis Targeting Chimeras) or molecular glues, provide mechanisms for achieving selective depletion of specific V-ATPase subpopulations. By conjugating V-ATPase-binding molecules to E3 ligase recruiters, researchers could develop agents that trigger ubiquitination and proteasomal degradation of V-ATPase complexes in specific cellular contexts. The modular nature of these approaches allows adjustment of tissue specificity through variation in the targeting ligand component.

Combination therapies targeting multiple aspects of pH dysregulation may provide synergistic benefits in complex disorders. By pairing V-ATPase modulators with agents addressing downstream consequences of pH disturbances (such as altered enzyme activities, protein misfolding, or ion channel function), more comprehensive correction of disease phenotypes may be achieved. Preclinical testing in rat models with manipulated Atp6v0e1 expression can validate these combinatorial approaches before translation to human applications.

How will systems biology approaches enhance our understanding of Atp6v0e1 in cellular pH homeostasis networks?

Systems biology approaches are poised to transform our understanding of Atp6v0e1's role within broader cellular pH homeostasis networks. By integrating multi-omics data with computational modeling, researchers can contextualize V-ATPase functions within complex regulatory systems, revealing emergent properties and unexpected network interactions that influence cellular pH regulation across physiological and pathological states.

Multi-omics integration combining transcriptomics, proteomics, metabolomics, and lipidomics data from models with manipulated Atp6v0e1 can reveal cascading effects of V-ATPase dysfunction across cellular systems. This approach becomes particularly powerful when temporal sampling captures pathway activation sequences following acute Atp6v0e1 perturbation. For example, rAAV6-mediated expression or knockdown of Atp6v0e1 in specific cell populations followed by time-course multi-omics analysis can reveal primary versus secondary consequences of altered V-ATPase function .

Mathematical modeling of intracellular pH regulation incorporating V-ATPase dynamics provides predictive frameworks for understanding system behavior under various conditions. Ordinary differential equation models capturing the kinetics of major pH regulatory transporters (including V-ATPase, Na+/H+ exchangers, bicarbonate transporters) and buffer systems can simulate pH responses to physiological challenges or pharmacological interventions. These models become particularly valuable when parameterized with experimental data from rat systems with manipulated Atp6v0e1 levels, enabling in silico prediction of intervention outcomes before experimental validation.

Network analysis approaches can identify regulatory hubs and feedback mechanisms controlling V-ATPase function. By constructing interaction networks incorporating transcriptional regulators, signaling pathways, and post-translational modification enzymes affecting Atp6v0e1 and other V-ATPase components, researchers can identify critical control points within the system. These analyses often reveal non-obvious intervention targets that may produce more robust or specific effects than directly targeting V-ATPase components.

Flux balance analysis incorporating V-ATPase-dependent processes can predict metabolic consequences of altered Atp6v0e1 function. Since intracellular pH affects numerous enzymatic reactions, changes in V-ATPase activity have far-reaching metabolic implications. Constraint-based modeling approaches can predict how alterations in organelle acidification affect metabolic pathway activities, providing insights into systemic consequences of V-ATPase dysfunction and potentially identifying metabolic vulnerabilities that could be therapeutically targeted.

Multi-scale modeling integrating molecular, cellular, and tissue-level processes provides comprehensive frameworks for understanding V-ATPase functions in complex physiological contexts. By connecting molecular simulations of Atp6v0e1 within V-ATPase complexes to cellular models of pH regulation and further to tissue-level acid-base balance, researchers can track how molecular perturbations propagate across scales. These approaches become especially valuable for understanding disorders affecting tissues with specialized pH regulation requirements, such as brain, kidney, or bone, where cell-type-specific expression of Atp6v0e1 may contribute to differential vulnerability to pH disturbances.

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.