ATP synthase subunit g (ATP5L) is encoded by the ATP5MG gene in humans and is part of the mitochondrial ATP synthase’s Fo complex. While its exact biochemical role remains unclear, subunit g is essential for ATP synthase dimerization and stability . ATP5L antibodies enable researchers to:
Detect ATP5L expression in tissues (e.g., liver, kidney) and cell lines (e.g., HeLa, HepG2) .
Investigate mitochondrial dysfunction in diseases like cardiomyopathy and metabolic disorders .
ATP5L antibodies are produced in rabbits and validated for applications including Western blot (WB), immunoprecipitation (IP), and immunohistochemistry (IHC). Key features are summarized below:
Subunit g is critical for ATP synthase dimerization. Knockout of Atp5l in mice disrupts oligomer formation, reducing mitochondrial membrane potential and sensitizing cells to necrosis .
Key finding: Loss of ATP5L exacerbates myocardial infarction size in ischemia-reperfusion models due to enhanced mitochondrial permeability transition pore (mPTP) opening .
ATP5L antibodies identified reduced ATP synthase activity in medulla oblongata tissues of hypertensive rats, linking mitochondrial dysfunction to cardiovascular pathology .
In mitochondrial diseases, ATP5L expression correlates with ATP synthase oligomerization and cell survival .
Model: Cardiomyocyte-specific Atp5l knockout mice exhibited:
Mechanism: ATP synthase deficiency sensitizes mPTP opening, increasing necrotic cell death .
ATP5L (ATP5MG) is a membrane subunit g of the mitochondrial ATP synthase complex, which plays a crucial role in ATP production. In humans, it has a length of 103 amino acid residues with a molecular weight of approximately 11.4 kDa . The protein is localized to the mitochondria, specifically the inner mitochondrial membrane . It functions as part of the F0 domain of ATP synthase, which forms the membrane proton channel .
ATP5L antibodies come in various forms with distinct characteristics:
Most ATP5L antibodies are validated for Western blotting applications, with dilution recommendations typically ranging from 1:500 to 1:2000 . It's worth noting that some antibodies may cross-react with the related protein ATP5L2 (ATP5MGL), which should be considered when interpreting experimental results .
Optimal Western blotting protocols for ATP5L antibodies typically follow these methodological steps:
Sample preparation: Prepare cell or tissue lysates using standard lysis buffers containing protease inhibitors to prevent degradation of the target protein.
Protein loading: Load 20-30 μg of total protein per lane . Due to the small size of ATP5L (11.4 kDa), using higher percentage (15%) SDS-PAGE gels is recommended for better resolution of low molecular weight proteins .
Gel electrophoresis and transfer: After SDS-PAGE separation, transfer proteins to PVDF or nitrocellulose membranes using standard transfer protocols.
Blocking: Block membranes with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.
Primary antibody incubation: Dilute ATP5L antibody according to manufacturer recommendations, typically in the range of 1:500-1:2000 . For validated antibodies, specific dilutions have been established:
Secondary antibody incubation: Use appropriate HRP-conjugated secondary antibody (typically anti-rabbit IgG) at dilutions of 1:1000-1:10,000 . Incubate for 1 hour at room temperature.
Detection: Visualize using standard ECL or other chemiluminescent detection methods.
It's important to note that the observed molecular weight may not always match the expected 11.4 kDa due to post-translational modifications or protein behavior in SDS-PAGE . Some researchers have reported observing multiple bands, which could represent different isoforms or modified versions of the protein.
Properly designed controls are critical for ensuring reliable and interpretable results when using ATP5L antibodies:
Positive controls:
Human cell lines with known ATP5L expression, such as HeLa cells
Human tissue lysates with high mitochondrial content, such as liver, heart, or kidney
293T cells and human fetal liver tissue have been verified as positive controls for Western blot applications
Negative controls:
Primary antibody omission control to assess non-specific binding of secondary antibody
Isotype control antibody (same species and isotype as the primary antibody but not reactive to the target)
When available, ATP5L knockout or knockdown samples
Peptide blocking control:
Cross-reactivity assessment:
If studying ATP5L specifically, verify that your antibody doesn't cross-react with ATP5L2/ATP5MGL by using recombinant proteins of both variants
For immunohistochemistry and immunofluorescence applications, include tissues known to express high levels of mitochondria as positive controls, and consider mitochondrial co-localization studies to confirm the expected subcellular localization pattern.
ATP5L antibodies offer valuable tools for investigating mitochondrial dysfunction in various disease contexts:
Assessment of ATP synthase complex integrity: ATP5L antibodies can be used in co-immunoprecipitation experiments to study interactions between ATP5L and other subunits of the ATP synthase complex. Changes in these interactions might indicate dysfunction in complex assembly.
Quantification of ATP5L expression levels: Altered expression of ATP synthase components, including ATP5L, has been associated with various pathological conditions. Western blotting with ATP5L antibodies can quantify these changes in disease models .
Subcellular localization studies: Immunofluorescence with ATP5L antibodies can reveal changes in the distribution pattern of ATP5L within cells, potentially indicating mitochondrial fragmentation or other structural abnormalities .
Blue Native PAGE analysis: When combined with other subunit-specific antibodies, ATP5L antibodies can help assess the integrity and assembly state of ATP synthase complexes in native gel electrophoresis.
Tissue distribution studies: Immunohistochemistry with ATP5L antibodies can reveal tissue-specific changes in mitochondrial content or ATP synthase expression in disease states .
When designing such studies, it's important to include appropriate controls and consider using multiple antibodies targeting different subunits of the ATP synthase complex to gain a comprehensive understanding of mitochondrial function.
Differentiating between ATP5L and its homolog ATP5L2 (ATP5MGL) requires careful experimental design and antibody selection:
Specific antibodies: Some antibodies are reported to recognize both ATP5L and ATP5L2 , while others may be specific to one isoform. Carefully review the antibody specifications and validation data to select one with the appropriate specificity.
Immunogen sequence analysis: Compare the immunogen sequence used to generate the antibody with the sequences of both ATP5L and ATP5L2. Antibodies raised against regions with higher sequence divergence are more likely to distinguish between the homologs.
Recombinant protein controls: Use purified recombinant ATP5L and ATP5L2 proteins as controls in Western blot experiments to determine cross-reactivity.
siRNA/shRNA knockdown validation: Perform selective knockdown of either ATP5L or ATP5L2 and observe the effect on antibody signal to determine specificity.
Mass spectrometry confirmation: For critical experiments, consider following up antibody-based detection with mass spectrometry analysis to unambiguously identify the detected protein.
Tissue-specific expression patterns: ATP5L and ATP5L2 may have different tissue expression patterns that can help distinguish between them in certain experimental contexts.
Observing unexpected band sizes with ATP5L antibodies is a common issue with several potential explanations:
Post-translational modifications: Although the calculated molecular weight of ATP5L is approximately 11 kDa, modifications such as phosphorylation, glycosylation, or ubiquitination can increase the apparent molecular weight .
Incomplete denaturation: The hydrophobic nature of membrane proteins like ATP5L can lead to incomplete denaturation, causing aberrant migration patterns.
Protein aggregation: ATP5L may form homo-oligomers or aggregates that are not fully disrupted by standard SDS-PAGE conditions.
Cross-reactivity: Some ATP5L antibodies may cross-react with related proteins, including ATP5L2 or other ATP synthase subunits .
Proteolytic processing: ATP5L may undergo proteolytic processing during mitochondrial import or as part of its normal lifecycle.
Sample preparation issues: Inadequate sample preparation can lead to degradation products or artifactual bands.
As noted in the product information for some commercial antibodies, "The actual band is not consistent with the expectation. Western blotting is a method for detecting a certain protein in a complex sample based on the specific binding of antigen and antibody. Different proteins can be divided into bands based on different mobility rates. The mobility is affected by many factors, which may cause the observed band size to be inconsistent with the expected size."
To address this issue, consider using positive controls with verified ATP5L expression, optimizing sample preparation protocols, and validating results with alternative techniques or additional antibodies.
Validating the specificity of ATP5L antibody signals is crucial for generating reliable scientific data. Key considerations include:
Multiple antibodies approach: Use multiple antibodies targeting different epitopes of ATP5L to confirm consistent results.
Genetic validation: Employ siRNA/shRNA knockdown or CRISPR/Cas9 knockout models to confirm the specificity of the signal. A reduction or absence of signal in these models strongly supports antibody specificity.
Peptide competition assays: Pre-incubate the antibody with its immunizing peptide before application to samples. Specific signals should be significantly reduced or eliminated.
Correlation with mRNA levels: Compare protein expression patterns with mRNA expression data from RT-PCR or RNA-seq.
Subcellular localization consistency: For immunohistochemistry or immunofluorescence, confirm that the observed localization pattern matches the expected mitochondrial distribution, potentially using co-staining with established mitochondrial markers.
Species cross-reactivity validation: If using the antibody across multiple species, validate specificity separately for each species, as epitope conservation may vary.
Mass spectrometry validation: For critical applications, consider immunoprecipitation followed by mass spectrometry to confirm that the antibody is indeed capturing ATP5L.
Positive and negative control tissues/cells: Include samples known to express high levels of ATP5L (e.g., human fetal liver, 293T cells) and those with minimal expression .
Implementing these validation strategies will significantly increase confidence in experimental results and help address the challenge of antibody specificity that affects many research areas.
Recent advances in active learning strategies for antibody-antigen binding prediction have potential applications for ATP5L antibody research. Active learning can reduce the costs of experimental binding data generation by starting with a small labeled subset and iteratively expanding the dataset .
This approach is particularly valuable for ATP5L antibodies given:
Epitope mapping optimization: Active learning algorithms can help identify the most informative epitope variants to test, potentially reducing the number of required antigen mutant variants by up to 35% .
Cross-reactivity prediction: These methods can accelerate the identification of potential cross-reactivity between ATP5L antibodies and related proteins like ATP5L2.
Optimization of library-on-library screening: For researchers developing new ATP5L antibodies, active learning approaches can improve the efficiency of library-on-library screening by prioritizing the most informative antibody-antigen pairs to test.
Out-of-distribution prediction: Machine learning models employing active learning can predict antibody-antigen interactions even when test antibodies and antigens are not represented in the training data .
Implementing these approaches requires:
Initial small-scale binding data for ATP5L antibodies
Computational infrastructure for running active learning algorithms
Iterative experimental validation
According to recent research, three of fourteen tested active learning algorithms significantly outperformed random data labeling approaches, speeding up the learning process by 28 steps compared to random baselines . This suggests that similar approaches could substantially improve the efficiency of ATP5L antibody characterization and development.
Recent advances in predicting antibody developability using molecular surface descriptors have important implications for developing effective ATP5L antibodies:
Molecular surface descriptors specifically designed for antibody developability can predict crucial biophysical properties including:
Viscosity prediction: High viscosity can complicate antibody formulation and delivery, particularly important for therapeutic applications.
Aggregation propensity: ATP5L antibodies with high aggregation propensity may show reduced specificity and reliability in experimental applications.
Hydrophobic interaction profiling: Surface hydrophobicity patterns can significantly influence an antibody's behavior in various buffer conditions.
Polyspecificity assessment: Molecular descriptors can help predict whether an ATP5L antibody might bind non-specifically to other targets.
Research has shown that averaging descriptor values over conformational distributions from molecular dynamics simulations can mitigate systematic shifts across different structure prediction methods . Six in silico developability risk flags have been proposed that can assess potential developability issues for candidate molecules .
For researchers developing or selecting ATP5L antibodies, considering these molecular surface descriptors could help:
Select antibodies with optimal biophysical properties
Predict potential issues before experimental validation
Guide optimization of existing antibodies
The sensitivity of these surface descriptors to methodological variables (such as dielectric constant choice, hydrophobicity scales, and structure prediction methods) should be carefully considered when applying these approaches to ATP5L antibody development .
ATP5L antibodies have significant potential for advancing our understanding of mitochondrial dynamics in neurodegenerative diseases:
Mitochondrial fragmentation analysis: ATP5L antibodies can be used in immunofluorescence studies to visualize changes in mitochondrial morphology and distribution in neuronal models of diseases like Alzheimer's, Parkinson's, and Huntington's.
ATP synthase dimerization studies: Recent research suggests that ATP synthase dimerization plays a role in mitochondrial cristae formation. ATP5L antibodies could help investigate whether this process is altered in neurodegenerative conditions.
Post-translational modification profiling: Using ATP5L antibodies in combination with modification-specific antibodies could reveal disease-specific changes in post-translational modifications of ATP synthase components.
Mitochondrial permeability transition pore (mPTP) research: The ATP synthase complex has been implicated in forming the mPTP, which plays a role in cell death. ATP5L antibodies could help investigate structural changes in the complex during mPTP formation in neurodegeneration.
Mitophagy assessment: ATP5L antibodies can be used to track the fate of mitochondria during mitophagy, a process often dysregulated in neurodegenerative diseases.
Future research should focus on developing more specific ATP5L antibodies that can distinguish between different conformational states of the protein, potentially revealing dynamic changes in ATP synthase structure during disease progression.
Several methodological advances could significantly improve the specificity and sensitivity of ATP5L antibodies:
Epitope-focused antibody development: Generating antibodies against unique epitopes of ATP5L that have minimal sequence homology with ATP5L2 and other related proteins would improve specificity.
Conformation-specific antibodies: Developing antibodies that recognize specific conformational states of ATP5L within the ATP synthase complex could provide insights into dynamic structural changes.
Site-specific modification detection: Creating antibodies that specifically recognize post-translationally modified forms of ATP5L would enable more detailed studies of its regulation.
Nanobody and single-domain antibody approaches: These smaller antibody formats may access epitopes that are sterically hindered in the assembled ATP synthase complex.
Multiplexed antibody validation: Establishing comprehensive validation pipelines that combine multiple orthogonal techniques (proteomics, genetic knockouts, etc.) would improve confidence in antibody specificity.
Standardized reporting of validation data: More complete disclosure of validation experiments, including all negative results, would help researchers select the most appropriate antibodies for their specific applications.
Application-specific optimization: Developing and sharing optimized protocols for specific applications (WB, IHC, IF, IP, etc.) would improve reproducibility across laboratories.
Advances in these areas would not only benefit ATP5L research specifically but could also serve as a model for improving antibody quality across the field of mitochondrial research.