ATP1B3 is the beta-3 subunit of Na+/K+-ATPase, encoded by the ATP1B3 gene in humans. It belongs to the family of Na+/K+ and H+/K+ ATPases beta chain proteins, which function as plasma membrane pumps with numerous physiological roles . The gene is mapped to chromosome 3q23, with a pseudogene located on chromosome 2 .
The protein consists of 279 amino acids with a single transmembrane domain and functions as the non-catalytic component of the active enzyme . The C-terminal portion of ATP1B3 folds into an immunoglobulin-like domain, which is critical for its structural and functional properties .
Key characteristics of ATP1B3:
Feature | Description |
---|---|
Protein Name | Sodium/potassium-transporting ATPase subunit beta-3 |
Gene Name | ATP1B3 |
UniProt ID | P54709 |
Transmembrane Domains | 1 |
Length | 279 amino acids |
Chromosome Location | 3q23 |
ATP1B3 serves as a key component of the Na+/K+-ATPase complex, which is essential for establishing and maintaining electrochemical gradients of Na+ and K+ ions across the plasma membrane . This function is critical for:
Osmoregulation in various cell types
Sodium-coupled transport of organic and inorganic molecules
Maintaining electrical excitability of nerve and muscle cells
Additionally, ATP1B3 has been implicated in immune function, with research demonstrating its ability to up-regulate lymphocyte activity and promote the production of several cytokines including IFN-γ, IL-2, IL-4, and IL-10 . This suggests ATP1B3 may function beyond its classical role in ion transport to influence immune responses.
Recent studies also indicate ATP1B3 acts as a co-factor that accelerates BST-2 degradation and reduces BST-2-mediated restriction of HIV-1 production and NF-κB activation, suggesting a role in viral infection dynamics .
ATP1B3 shows differential expression patterns across human tissues. While the search results don't provide comprehensive details on all tissues, they reveal important patterns in specific tissues such as the cochlea, where ATP1B3 transcript distribution has been mapped .
In the cochlea, ATP1B3 expression appears to be cell-type specific. Type I spiral ganglion cells express ATP1B1 gene transcripts at levels exceeding those in marginal cells by approximately 20 times and type II fibrocytes by nine times . Interestingly, axons have been found to lack ATP1B3 gene transcripts, while the NKAβ1 protein is richly expressed along the axonal plasma membrane in certain neuronal populations .
The regulation of ATP1B3 expression appears to be context-dependent, with evidence of altered expression in various pathological states, including cancer. This suggests complex transcriptional and post-transcriptional regulatory mechanisms that warrant further investigation.
ATP1B3 has emerged as a significant factor in cancer biology, with evidence supporting its oncogenic role in multiple cancer types:
ATP1B3 expression correlates positively with markers of several immune cell populations in the tumor microenvironment, including:
This correlation with immune infiltration has been validated at both mRNA and protein levels using proteomics datasets .
In gastric cancer, ATP1B3 overexpression promotes multiple aspects of cancer progression, including:
Mechanistically, ATP1B3 appears to promote malignant progression in gastric cancer through activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway .
In glioma, ATP1B3 may influence tumor cell proliferation and migration through modulation of the MAPK and NF-κB signaling pathways . Knockdown experiments in glioma cell lines (U87MG and U251MG) have demonstrated that reducing ATP1B3 expression impacts the invasive and proliferative capacities of these cells .
Based on the search results, several methodological approaches have proven effective for investigating ATP1B3 function:
Gene Expression Analysis:
Functional Knockdown Studies:
Phenotypic Assays:
Protein-Protein Interaction Studies:
Signaling Pathway Analysis:
Western Blotting to assess changes in signaling pathway components following ATP1B3 manipulation
Analysis of phosphorylation status of key signaling molecules in the MAPK pathway (p-Raf1, p-MEK 1/2, p-ERK 1/2) and NF-κB pathway (p-IκBα, p-P65)
Investigation of downstream effectors such as Cyclin D1 and VEGFA
Integrative Approaches:
ATP1B3 expression demonstrates significant correlations with patient outcomes across multiple cancer types, though the prognostic implications appear to be cancer-specific:
In hepatocellular carcinoma (HCC):
In gliomas:
Database analysis reveals a negative correlation between ATP1B3 expression and patient outcomes
Higher expression appears to correlate with more aggressive disease
In gastric cancer:
Overexpression of ATP1B3 is associated with adverse clinical outcomes
The protein promotes tumor cell proliferation, invasion, anti-apoptosis mechanisms, and cell-cycle dysregulation
These effects appear to be mediated through the PI3K/AKT signaling pathway
The studies collectively suggest that ATP1B3 overexpression is generally associated with poorer prognosis across multiple cancer types, making it a potential biomarker for disease progression and treatment response.
Several technical challenges exist in ATP1B3 research that researchers should be aware of:
Distinguishing between ATP1B family members:
The ATP1B family includes multiple members (ATP1B1, ATP1B2, ATP1B3, ATP1B4) with partially overlapping functions. Designing highly specific primers, antibodies, and siRNAs is crucial to avoid cross-reactivity. Validation using multiple methods (qPCR, Western blot, immunostaining) is recommended to ensure specificity.
Subcellular localization assessment:
As a membrane protein with a single transmembrane domain, properly preserving membrane structures during sample preparation is essential. For immunofluorescence studies, appropriate permeabilization protocols that maintain membrane integrity while allowing antibody access are necessary .
Functional redundancy:
Other Na+/K+-ATPase β subunits may compensate for ATP1B3 knockdown. Consider using combinatorial knockdown approaches or rescue experiments to address this issue.
Protein-protein interaction studies:
The interaction partners of ATP1B3 (such as PPP1CA in glioma) may be tissue or context-specific . Use proximity ligation assays or FRET in addition to traditional co-immunoprecipitation to validate interactions in their native cellular context.
Translating in vitro findings to in vivo contexts:
Establish appropriate animal models that recapitulate the physiological or pathological context being studied. Consider conditional knockout/knockin models to avoid developmental effects of ATP1B3 manipulation.
When investigating ATP1B3 in cancer models, several essential controls should be implemented:
Expression controls:
Knockdown/overexpression controls:
Functional assay controls:
Perform rescue experiments by re-expressing siRNA-resistant ATP1B3 constructs
Include positive controls for cell proliferation, migration, and apoptosis assays
Time-course analyses to determine the temporal dynamics of ATP1B3-mediated effects
Signaling pathway controls:
Clinical correlation controls:
Given the emerging role of ATP1B3 in immune modulation, particularly in cancer contexts, several methodological approaches can effectively assess its immunomodulatory functions:
Immune cell infiltration analysis:
Cytokine profiling:
Co-culture systems:
Establish co-culture models of cancer cells and immune cells (T cells, macrophages, B cells)
Manipulate ATP1B3 expression in cancer cells and assess changes in immune cell activation, proliferation, and effector functions
Use transwell systems to distinguish between contact-dependent and secreted factor-mediated effects
Single-cell analysis:
Apply single-cell RNA sequencing to characterize heterogeneity in ATP1B3 expression and its correlation with immune phenotypes
Use CyTOF or spectral flow cytometry to simultaneously assess multiple immune parameters
Integrate these data with spatial information using techniques like imaging mass cytometry
In vivo immune monitoring:
Utilize syngeneic mouse models with ATP1B3 manipulation in cancer cells
Monitor changes in tumor-infiltrating lymphocytes, myeloid populations, and systemic immune parameters
Assess response to immunotherapies in the context of ATP1B3 modulation
Several promising therapeutic approaches targeting ATP1B3 in cancer warrant further investigation:
RNA interference-based therapies:
Small molecule inhibitors:
Design of specific inhibitors targeting the interaction between ATP1B3 and alpha subunits
Development of compounds that disrupt ATP1B3's interactions with downstream signaling partners
Repurposing of existing Na+/K+-ATPase inhibitors with optimized specificity for ATP1B3-containing complexes
Immunotherapeutic approaches:
Targeting downstream pathways:
Biomarker development:
Despite significant progress in understanding ATP1B3 function, several unexplored aspects merit further investigation:
Post-translational modifications:
The regulation of ATP1B3 through phosphorylation, glycosylation, or other modifications remains poorly characterized. Investigations into how these modifications affect ATP1B3 function, localization, and interactions could reveal new regulatory mechanisms.
Non-canonical functions:
Beyond its role in Na+/K+-ATPase activity, ATP1B3 may have additional functions, particularly in immune modulation and viral infection resistance . Systematic interactome studies could uncover novel binding partners and functional roles.
Tissue-specific roles:
While ATP1B3's functions have been studied in certain contexts (cancer, cochlea), its tissue-specific roles across different organ systems remain largely unexplored . Comparative studies across tissues could reveal specialized functions.
Developmental biology:
The role of ATP1B3 during embryonic development and in stem cell biology has received limited attention. Studies on ATP1B3's contribution to developmental processes and tissue differentiation could provide valuable insights.
Extracellular vesicle involvement:
Given its membrane localization, ATP1B3 may be incorporated into extracellular vesicles, potentially mediating intercellular communication. Investigating ATP1B3's presence and function in exosomes could reveal novel mechanisms of action.
Metabolic influences:
The relationship between ATP1B3 and cellular metabolism, particularly in the context of cancer's metabolic reprogramming, remains to be elucidated. Studies on how ATP1B3 affects or responds to metabolic states could uncover new therapeutic opportunities.
ATP1B3 research stands to benefit significantly from integration with emerging technologies:
CRISPR-based approaches:
Application of CRISPR screening to identify synthetic lethal interactions with ATP1B3
Development of CRISPR-based ATP1B3 regulators for precise temporal control
Use of base editing or prime editing for introducing specific ATP1B3 mutations
Spatial transcriptomics and proteomics:
Artificial intelligence and machine learning:
Development of predictive models for ATP1B3-associated patient outcomes
Integration of multi-omics data to uncover ATP1B3 regulatory networks
Virtual screening for ATP1B3-targeting compounds
Organoid technologies:
Establishment of patient-derived organoids to study ATP1B3 in more physiologically relevant systems
Development of co-culture organoid models incorporating immune components
High-throughput drug screening in ATP1B3-manipulated organoids
Single-molecule imaging:
Visualization of ATP1B3 dynamics in living cells using techniques like PALM/STORM
Analysis of ATP1B3 clustering and interaction with other membrane proteins
Real-time monitoring of ATP1B3's role in membrane organization
Nanobody and aptamer development:
Creation of highly specific ATP1B3-targeting nanobodies for functional studies
Development of aptamer-based sensors for monitoring ATP1B3 expression in real-time
Targeted delivery of therapeutic agents to ATP1B3-overexpressing cells
ATPase Transporting Beta 3, also known as ATP1B3, is a protein that plays a crucial role in the function of the sodium/potassium-transporting ATPase enzyme. This enzyme is essential for maintaining the electrochemical gradients of sodium (Na+) and potassium (K+) ions across the plasma membrane, which are vital for various cellular processes, including osmoregulation, sodium-coupled transport, and electrical excitability of nerve and muscle cells .
ATP1B3 is a non-catalytic component of the active enzyme complex, which catalyzes the hydrolysis of ATP coupled with the exchange of Na+ and K+ ions across the plasma membrane . The enzyme complex is composed of two subunits: a large catalytic subunit (alpha) and a smaller glycoprotein subunit (beta). ATP1B3 belongs to the family of Na+/K+ and H+/K+ ATPases beta chain proteins and is specifically a part of the Na+/K+ -ATPases subfamily .
The beta subunit, including ATP1B3, is responsible for the assembly of alpha/beta heterodimers, which control the quantity of sodium pumps transported to the plasma membrane . This regulation is crucial for maintaining the proper function and distribution of the sodium/potassium pumps.
ATP1B3 Human Recombinant is produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain containing 246 amino acids (57-279) and has a molecular mass of 27.4 kDa . The recombinant protein is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques .
The sodium/potassium-transporting ATPase, which includes ATP1B3, is integral to various physiological processes. These include:
Mutations or dysregulation of ATP1B3 and the sodium/potassium-transporting ATPase can lead to various medical conditions, including cardiac and neurological disorders. Understanding the function and regulation of ATP1B3 is essential for developing therapeutic strategies for these conditions.