The DARS2 Antibody (Catalog Number: 13807-1-AP) is a rabbit polyclonal antibody developed by Proteintech, designed to target the mitochondrial aspartyl-tRNA synthetase (DARS2) protein. This antibody is primarily used in research applications such as Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF/ICC), and enzyme-linked immunosorbent assay (ELISA). Its specificity for human, mouse, and rat samples makes it a versatile tool for cross-species studies .
Western Blotting: Detects DARS2 in lysates of mitochondrial fractions, confirming protein expression levels.
Immunohistochemistry: Localizes DARS2 in tissue sections, such as bladder cancer or brain samples .
Immunofluorescence: Visualizes mitochondrial DARS2 in cell cultures, aiding in subcellular localization studies .
DARS2 has been identified as a key regulator of bladder cancer progression. Studies using this antibody in IHC demonstrated elevated DARS2 expression in BLCA tissues compared to normal epithelium, correlating with poor prognosis . Its role in mitophagy regulation and cell cycle progression highlights its potential as a therapeutic target.
Mutations in DARS2 cause LBSL, a rare neurological disorder. The antibody has been used to study aberrant protein expression in patient-derived cerebral organoids, revealing dysregulated mitochondrial function and alternative splicing .
TIMER2 analysis showed that DARS2 expression inversely correlates with immune cell infiltration (e.g., B cells, CD4+ T cells) in bladder cancer, suggesting its role in shaping the tumor microenvironment .
The antibody’s specificity has been validated in multiple studies:
Western Blot: Detects a ~74 kDa band corresponding to DARS2 in mitochondrial lysates .
IHC: Exhibits strong staining in tumor tissues (e.g., BLCA, LUAD) with minimal background noise .
ELISA: Used to quantify DARS2 levels in patient sera for diagnostic purposes .
| Assay | Protocol Steps |
|---|---|
| WB | 1. Load 30 μg lysate/lane. 2. Use 1:1,000 dilution. 3. Detect with HRP-conjugated secondary. |
| IHC | 1. Paraffin sections. 2. Use 1:200 dilution. 3. Stain with DAB and counterstain with hematoxylin. |
| IF | 1. Fix cells with 4% PFA. 2. Use 1:500 dilution. 3. Visualize with Alexa 488-conjugated secondary. |
DARS2 antibodies have been successfully validated for several key applications in research, with varying optimal dilution ratios:
| Application | Recommended Dilution Range | Validated Cell/Tissue Types |
|---|---|---|
| Western Blot (WB) | 1:1000-1:4000 | K-562 cells, human placenta tissue, U-937 cells |
| Immunohistochemistry (IHC) | 1:50-1:500 | Mouse cerebellum, human brain tissue, human testis tissue, human gliomas tissue |
| Immunofluorescence (IF/ICC) | 1:50-1:500 | A431 cells |
These applications allow researchers to evaluate DARS2 expression, localization, and protein interactions in various experimental contexts. For Western blot applications, DARS2 typically appears at 66-74 kDa, with some published studies reporting bands at 55 kDa, 50 kDa, or 66 kDa . When working with new cell lines or tissues, it is advisable to first optimize antibody concentrations through dilution series experiments.
Most commercial DARS2 antibodies are supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3. For long-term storage, maintain at -20°C where they typically remain stable for at least one year after shipment. For smaller volume antibodies (20μl), manufacturers often include 0.1% BSA as a stabilizer .
Important handling considerations:
Avoid repeated freeze-thaw cycles by aliquoting upon receipt
Bring to room temperature before opening to prevent condensation
Centrifuge briefly before use to collect solution at the bottom of the tube
Return promptly to -20°C after use
For diluted working solutions, prepare fresh for each experiment or store at 4°C for no longer than one week
Validating DARS2 antibody specificity requires careful experimental design to avoid cross-reactivity and false positives. The most rigorous approach involves:
Positive controls: Use tissues/cells with known DARS2 expression (e.g., K-562 cells, human placenta tissue)
Negative controls: Include DARS2 knockdown or knockout samples alongside test samples
Multiple detection methods: Validate findings using at least two techniques (e.g., WB and IHC)
Peptide competition assays: Pre-incubate antibody with immunizing peptide to confirm specificity
Multiple studies have used siRNA-mediated DARS2 knockdown to validate antibody specificity, demonstrating reduced antibody signal in Western blot and immunohistochemistry applications . This approach provides convincing evidence for antibody specificity while simultaneously allowing functional studies of DARS2 depletion.
Research demonstrates that DARS2 expression is significantly altered in multiple cancer types, with distinct patterns across different malignancies:
For bladder cancer specifically, DARS2 upregulation correlates with tumor progression and poor prognosis . Functional studies demonstrate that DARS2 knockdown inhibits cancer cell proliferation, metastasis, and tumorigenesis . When designing experiments to assess DARS2's role in cancer, researchers should include corresponding normal tissue controls and stratify samples by tumor stage and grade to properly evaluate expression patterns.
DARS2 expression correlates distinctly with immune cell infiltration patterns across different cancer types:
In bladder cancer:
Negative correlation with immune-active cells: CD4+ T cells (R= -0.251, P < 0.001) and NK cells (R= -0.067, P < 0.001)
Positive correlation with immunosuppressive cells: MDSCs (R= 0.372, P < 0.001) and macrophages (R= 0.196, P < 0.01)
Positive correlation with CD8+ T cells (R= 0.203, P < 0.001)
Positive correlation with PD-L1 expression (R= 0.202, P < 0.001)
In lung adenocarcinoma:
Negative correlation with B cells (r = -0.244, P = 5.32e-8)
Negative correlation with CD4+ T cells (r = -0.206, P = 4.91e-6)
Negative correlation with dendritic cells (r = -0.143, P = 1.55e-3)
When designing immunology-focused experiments, consider including co-staining for DARS2 and immune cell markers to validate these correlations in your specific experimental model. Flow cytometry and multiplex immunohistochemistry approaches are particularly valuable for characterizing these relationships.
DARS2 mutations cause leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), a rare neurological disorder. Key aspects of DARS2 in LBSL include:
Most LBSL patients (88%) carry a mutation in the intron 2 splice acceptor region, with one specific mutation (c.228-20_21delTTinsC) having a carrier frequency of 1:95 in the Finnish population
This mutation affects exon 3 splicing, causing a frameshift (p.R76SfsX5) and premature stop codon
The mutation is "leaky," allowing some production of functional mt-AspRS protein
Neural cells, particularly neurons, show profound splicing defects in this region, significantly reducing functional mt-AspRS levels
When studying DARS2 in neurological contexts, researchers should consider tissue-specific expression patterns and employ neuron-specific cell models to recapitulate disease-relevant conditions. Additionally, evaluating both protein levels and enzymatic activity provides more comprehensive insights into DARS2's role in disease pathogenesis.
For rigorous DARS2 functional studies, implement these experimental controls:
For knockdown experiments:
Negative control siRNA/shRNA: Use non-targeting sequences with similar GC content
Multiple siRNA sequences: Employ at least two distinct DARS2-targeting sequences to rule out off-target effects
Rescue experiments: Re-express siRNA-resistant DARS2 to confirm phenotype specificity
Quantification controls: Measure knockdown efficiency by both qRT-PCR and Western blot
For overexpression experiments:
Empty vector control: Must match backbone of DARS2 expression vector
Tagged vs. untagged constructs: Compare to ensure tag doesn't interfere with function
Expression level monitoring: Use inducible systems to achieve physiologically relevant levels
Functional validation: Confirm increased aminoacylation activity using biochemical assays
Research has demonstrated that DARS2 knockdown significantly inhibits cell proliferation in lung adenocarcinoma cell lines (A549 and H1299) and bladder cancer cell lines, validating the effectiveness of this approach for functional studies .
DARS2 splicing analysis is particularly important given the splice site mutations in LBSL. For comprehensive splicing analysis:
Primer design strategy:
Design primers that anneal to exon-exon junctions (exons 2, 3, and 4) to differentiate between inclusion and exclusion of exon 3
Quantitative assessment methods:
RT-PCR with visualization on agarose gels for qualitative assessment
RT-qPCR with exon junction-specific primers for quantitative analysis
RNA-seq with junction reads analysis for genome-wide splicing context
Minigene assays to test specific splicing regulatory elements
Cell type considerations:
Compare splicing patterns across relevant cell types (neurons, glia, cancer cells) as splicing efficiency varies significantly between cell types
A study by Wang et al. using DTUrtle (v0.8.1) for differential transcript usage analysis and BRIE2 (v2.0.5) for differential spliced exon analysis demonstrated cell type-specific splicing patterns of DARS2 . This methodological approach provides a comprehensive framework for researchers investigating splicing variations.
Recent research suggests DARS2 may have non-canonical functions beyond aminoacylation. To investigate these:
Protein interaction studies:
Co-immunoprecipitation followed by mass spectrometry to identify novel binding partners
Proximity labeling methods (BioID, APEX) to catalog the protein neighborhood
Yeast two-hybrid screening for direct interaction partners
Subcellular localization analysis:
Super-resolution microscopy with mitochondrial and other organelle markers
Subcellular fractionation with Western blot analysis
Live-cell imaging with fluorescently tagged DARS2
Pathway analysis approaches:
Phosphoproteomics to identify signaling pathways affected by DARS2 perturbation
Transcriptomics to identify gene expression networks regulated by DARS2
Metabolomics to identify metabolic pathways affected by DARS2 activity
Recent studies suggest DARS2 may be involved in mitophagy regulation through interactions with PINK1, offering a novel research direction beyond its classical role . Additionally, research by Sauter et al. identified DARS2 missense mutations in regions conserved only in mammals, suggesting evolutionarily acquired supplementary functions that may be involved in disease pathology .
To investigate DARS2's role in immune regulation through PD-L1:
Expression correlation analysis:
Perform co-expression analysis in patient samples and cell lines
Use multivariate analysis to control for confounding factors
Employ multiplexed IHC to visualize co-localization in tissue sections
Mechanistic investigations:
Assess PD-L1 levels after DARS2 knockdown/overexpression by flow cytometry and Western blot
Examine transcriptional regulation using reporter assays and ChIP
Investigate protein stability using pulse-chase experiments and proteasome inhibitors
Functional immune assays:
Co-culture cancer cells with immune cells to measure functional consequences
Use immune cell killing assays with DARS2-modulated cancer cells
Analyze immune checkpoint blockade efficacy in DARS2-high versus DARS2-low models
Research has demonstrated that in bladder cancer, DARS2 expression positively correlates with PD-L1 expression (R= 0.202, P < 0.001), and knockdown experiments showed that DARS2 modulation affects PD-L1 levels . This suggests DARS2 may facilitate immune evasion, making it a potential predictive indicator for immune therapy responses.
For investigating DARS2's role in cell cycle regulation:
Cell cycle analysis approaches:
Flow cytometry with propidium iodide or DAPI staining
EdU incorporation assays for S-phase analysis
Time-lapse microscopy with cell cycle reporters (FUCCI system)
Cell cycle protein expression:
Western blot analysis of key regulators (CDK4, CDK6, p53, p21)
Immunofluorescence for spatial distribution of cell cycle markers
Proteomic analysis of cell cycle protein complexes
Functional assays:
Cell synchronization followed by release with DARS2 modulation
CDK activity assays to assess direct functional impact
Rescue experiments with cell cycle regulators
Research by Liu et al. found that DARS2 knockdown in bladder cancer cells reduced CDK4 expression while CDK6, p53, and p21 levels remained unchanged, suggesting DARS2 specifically promotes G1-to-S phase transition by upregulating CDK4 . This methodological approach provides a template for researchers investigating DARS2's role in cell cycle regulation across different cancer types.
For developing DARS2 as a clinical biomarker:
Biomarker validation approach:
Multi-cohort analysis with discovery and validation sets
Multivariate analysis controlling for clinical variables
Comparison with established biomarkers
Standardized cutoff determination using ROC analysis
Technical considerations:
Antibody validation across multiple platforms (IHC, ELISA, multiplex assays)
Standard operating procedures for specimen handling
Internal controls for normalization
Reproducibility testing across laboratories
Combined biomarker strategies:
Creation of multi-gene prognostic scores
Integration with established clinical parameters
Machine learning approaches for biomarker panel optimization
Research has demonstrated the potential of DARS2 as a prognostic biomarker, particularly when combined with other markers. In bladder cancer, researchers developed a DARS2-PINK1-CDK4 axis expression score that showed significant prognostic value across multiple datasets . This multi-gene approach improved prognostic accuracy compared to DARS2 expression alone.
DARS2 has been reported with varying molecular weights across studies, creating potential confusion in data interpretation:
| Reported Molecular Weight | Study/Source | Contributing Factors |
|---|---|---|
| 74 kDa (predicted) | Multiple commercial sources | Full-length protein sequence prediction |
| 66-74 kDa | Proteintech antibody documentation | Post-translational modifications, mitochondrial targeting sequence |
| 55 kDa | Published literature | N-terminal processing of mitochondrial targeting sequence |
| 50 kDa | Published literature | Alternative splicing or proteolytic processing |
| 70 kDa | Abcam antibody documentation | Differences in gel systems or molecular weight markers |
To resolve discrepancies:
Include positive control samples with known DARS2 expression
Use multiple antibodies targeting different epitopes
Verify specificity through knockdown/knockout experiments
Consider pre-process mature mitochondrial DARS2 versus precursor forms
Note differences in rat DARS2, which has a ~40 amino acid N-terminal extension that can be removed without affecting catalytic activity
These variations may reflect biological differences in DARS2 processing or technical differences in experimental conditions rather than antibody specificity issues.
DARS2 shows divergent expression patterns across cancer types, with overexpression in some (bladder cancer, lung adenocarcinoma) and underexpression in others (kidney cancers, thyroid carcinoma) . To reconcile these differences:
Technical reconciliation:
Standardize analysis methods across studies
Verify antibody specificity in each tissue type
Use multiple detection methods (protein and mRNA)
Include appropriate tissue-specific controls
Biological interpretation:
Consider tissue-specific mitochondrial dependency
Analyze mitochondrial content differences between tissues
Examine tissue-specific splicing patterns
Investigate tissue-specific roles beyond aminoacylation
Experimental approach:
Perform systematic pan-cancer analysis with consistent methodology
Use paired normal-tumor samples for each cancer type
Stratify by molecular subtypes within each cancer type
Correlate with mitochondrial function metrics
Understanding these context-dependent patterns is crucial for developing DARS2-targeted therapeutic strategies and avoiding off-target effects in tissues where DARS2 levels are reduced in cancer.
To distinguish between canonical and non-canonical DARS2 functions:
Separation-of-function mutations:
Engineer mutations that specifically disrupt aminoacylation without affecting protein interactions
Compare phenotypes between complete knockout and function-specific mutants
Use domain deletion constructs to map functional regions
Biochemical activity assays:
Measure aminoacylation activity using in vitro assays
Correlate activity levels with disease phenotypes
Use aminoacylation inhibitors to phenocopy genetic approaches
Evolutionary analysis:
Focus on mutations in mammalian-specific conserved regions vs. universally conserved regions
Compare functions across species with different DARS2 domain architectures
Analyze disease mutations for their evolutionary conservation patterns
Sauter et al. identified missense mutations in DARS2 regions conserved only in mammals, suggesting evolutionarily acquired supplementary functions may contribute to LBSL pathology . Additionally, research on DARS2's role in mitophagy regulation through PINK1 pathway modulation points to non-canonical functions beyond aminoacylation .