ASNA1 antibodies are designed to bind specifically to the ASNA1 protein (also known as TRC40), enabling its detection in biological samples. These antibodies are categorized by their clonality, host species, and reactivity:
Monoclonal antibodies (e.g., [2A1]) offer high specificity but limited cross-reactivity.
Polyclonal antibodies (e.g., 15450-1-AP) provide broader epitope coverage and versatility across species.
Rabbit-derived antibodies are often preferred for Western blotting and immunohistochemistry due to strong signal amplification.
ASNA1 exhibits two distinct biochemical roles:
ASNA1 binds TAPs via its hydrophobic groove, delivering them to the endoplasmic reticulum (ER) for membrane insertion. This process is ATP-dependent and requires interaction with ER receptors like WRB/CAML .
In oxidized states, ASNA1 acts as a holdase, stabilizing misfolded proteins. This function is critical for stress responses and insulin secretion .
ASNA1 antibodies are employed across diverse experimental platforms:
C. elegans models showed that ASNA1 mutations (e.g., ΔHis164, A63V) sensitize cells to cisplatin without affecting insulin secretion, highlighting ASNA1 as a druggable target .
Human studies linked low ASNA1 expression to cisplatin resistance, suggesting its potential as a predictive biomarker .
In Eimeria tenella, ASNA1 overexpression correlates with resistance to diclazuril and salinomycin. Antibody inhibition reduced sporozoite invasion of host cells .
ASNA1 antibodies identified mutations (e.g., A149V) conferring resistance to Retro-2, a compound blocking retrograde transport. This confirmed ASNA1 as a key target in ricin toxin resistance .
ASNA1 (also known as TRC40) is a highly conserved ATPase initially characterized for its role in arsenite and antimonite efflux systems. More recent research has established ASNA1 as a critical component of the transmembrane recognition complex (TRC) pathway, which mediates tail-anchored protein insertion into the endoplasmic reticulum membrane . The protein has a calculated molecular weight of 38.7 kDa, though it typically appears at 37-41 kDa in experimental analyses . ASNA1's ATPase activity is essential for proper pancreatic development, with knockout models demonstrating its requirement for epithelial tissue maintenance . The protein plays a particularly important role in the subcellular localization of TA-SNAREs such as Stx5/Sed5 and Stx6, which undergo significant relocalization from their normal positions in the cis-Golgi and trans-Golgi network respectively when ASNA1 function is compromised .
ASNA1 antibodies have been validated for multiple experimental applications, each requiring specific optimization conditions. The validated applications include:
Application | Recommended Dilution | Positive Detection Examples |
---|---|---|
Western Blot (WB) | 1:500-1:1000 | Human brain tissue, HeLa cells, human heart tissue, mouse brain tissue |
Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg total protein | Mouse brain tissue |
Immunohistochemistry (IHC) | 1:20-1:200 | Human lung cancer tissue |
Immunofluorescence (IF)/ICC | 1:200-1:800 | HepG2 cells |
Antigen retrieval protocols significantly impact IHC results, with TE buffer (pH 9.0) being recommended, although citrate buffer (pH 6.0) may serve as an alternative . Each application requires careful titration in specific experimental systems for optimal signal-to-noise ratios, particularly when working with novel tissue samples or cellular models.
ASNA1 antibodies demonstrate extended stability when properly stored. The recommended storage conditions include maintaining the antibody at -20°C in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . Under these conditions, the antibody remains stable for one year post-shipment. Importantly, aliquoting is not necessary for -20°C storage, which differs from many other antibody storage protocols. When working with smaller quantities (20 μl sizes), researchers should note that these preparations contain 0.1% BSA as a stabilizer . Repeated freeze-thaw cycles should be minimized, and upon initial thawing, the antibody should be gently mixed by inversion rather than vortexing to prevent protein denaturation and maintain epitope recognition capacity.
ASNA1 antibodies serve as valuable tools for investigating protein trafficking pathways, particularly for tail-anchored membrane proteins. When combined with subcellular markers, these antibodies can reveal critical mechanisms of membrane protein insertion. For example, co-localization studies using ASNA1 antibodies alongside markers for the Golgi apparatus (such as GM130) have demonstrated that ASNA1 dysfunction results in striking relocalization of SNARE proteins including Stx5 and Stx6 from their normal positions in the cis-Golgi and trans-Golgi network .
Experimental approaches typically involve:
Fixing cells in 4% PFA for 10 minutes
Permeabilizing with 0.3% Triton-X for 10 minutes
Blocking in fresh 3% BSA for at least 1 hour
Incubating with primary antibodies for 1 hour (anti-ASNA1 at 1:300 dilution)
Applying secondary antibodies for 1 hour (anti-rabbit-647 at 1:2000)
Imaging using confocal microscopy with oil immersion (Plan Apo, NA-1.5, 60× or 100×)
For quantitative analysis, maximum projections from 0.2 μm Z-stacks should be generated using imaging software such as FIJI, with subsequent analysis performed using specialized imaging packages like csth and pyto segmenter .
Multiple genetic manipulation strategies have demonstrated efficacy in studying ASNA1 function:
CRISPR/Cas9 knockout: CRISPR-based approaches have established ASNA1 as an essential gene for cell growth in K562 cells . Complete knockout is typically lethal, necessitating conditional approaches for in vivo studies.
shRNA knockdown: Stable expression of shRNAs targeting ASNA1 has been successfully implemented in HeLa cells using lentiviral delivery systems. This approach allows for partial depletion and subsequent functional studies, including sensitivity to compounds like Retro-2 .
Point mutation generation: CRISPR-directed diversification strategies have identified functional mutations such as C460T (resulting in A149V amino acid change), which confers resistance to ASNA1 inhibitors like DHQZ36.1 . This approach involves:
Conditional knockout in animal models: Tissue-specific deletion using Cre-loxP systems has revealed developmental roles of ASNA1, such as the Asna1Panc−/− mouse model which develops pancreatic and duodenal epithelial hypoplasia .
For protein tagging and localization studies, N-terminal fusion of BFP to ASNA1 has been successfully implemented using Gibson assembly with a GGGS linker separating ASNA1 from BFP .
Several quantitative approaches have been validated for measuring ASNA1-dependent cellular phenotypes:
Cell viability/toxicity assays: ASNA1 knockdown sensitizes cells to growth inhibition by compounds like Retro-2 . Quantification protocols involve:
Plating cells at defined density (25,000 cells per well in 24-well plates)
Pretreatment with compound of interest for 1 hour
Treatment with toxin (e.g., 2.5 ng/mL ricin) for 24 hours
Media replacement with fresh compound and viability indicator (50 nM CellTox Green)
Time-lapse imaging every 4 hours for 72 hours using systems like Incucyte Zoom
Flow cytometry for protein trafficking: For quantification of ASNA1's role in membrane protein insertion, approaches using GFP-2A-RFP-SEC61BTMD reporter systems have been established:
Treatment of cells with doxycycline (250 ng/mL) for approximately 20 hours
Cell sorting for GFP and RFP positive populations
Transfection with vectors expressing BFP-ASNA1 variants
Recovery in reduced serum media for 1 hour before compound treatment
Flow cytometric analysis using appropriate laser configurations
Apoptosis quantification: In developmental contexts, quantification of cleaved caspase-3 positive cells and TUNEL assays have revealed ASNA1's role in preventing apoptosis, particularly in pancreatic epithelial cells .
Several technical challenges may arise when working with ASNA1 antibodies:
Antigen retrieval challenges in IHC: For formalin-fixed tissues, the choice of antigen retrieval buffer significantly impacts staining quality. While TE buffer (pH 9.0) is recommended, some tissues may require optimization with citrate buffer (pH 6.0) . When encountering weak or inconsistent staining, systematically compare both buffer systems, adjusting incubation times (10-20 minutes) and temperatures (95-100°C).
Background in immunofluorescence: When performing IF/ICC applications, high background can obscure specific signals. This can be addressed by:
Co-localization analysis challenges: When examining ASNA1's relationship with organelle markers like STX5 or GM130, signal bleed-through between channels can compromise data interpretation. Implement sequential scanning protocols during confocal microscopy, and validate findings using appropriate controls including secondary-only samples and single-labeled preparations .
Validating antibody specificity is crucial for generating reliable data. Multiple approaches have been established for ASNA1 antibodies:
Genetic validation: Utilizing ASNA1 knockdown (shRNA) or knockout (CRISPR) approaches provides the most definitive control for antibody specificity. The disappearance or significant reduction of signal following genetic ablation confirms specificity .
Peptide competition: Pre-incubating the antibody with the immunizing peptide (ASNA1 fusion protein Ag7713) should eliminate specific signal in all applications while leaving non-specific binding intact.
Cross-validation with multiple antibodies: When possible, compare results using antibodies raised against different epitopes of ASNA1.
Molecular weight verification: In Western blotting, ASNA1 should appear between 37-41 kDa, which aligns with its calculated molecular weight of 38.7 kDa . Significant deviations suggest potential cross-reactivity or degradation.
When investigating ASNA1 inhibition by compounds such as Retro-2 or DHQZ36.1, several controls are essential:
Dose-response relationships: Establish complete dose-response curves (typically 0.1-20 μM) to identify both sub-effective and potentially toxic concentrations. For example, high concentrations of DHQZ36.1 can be toxic to cell growth even in wildtype cells .
Genetic rescue experiments: Express ASNA1 variants (e.g., A149V mutant) resistant to inhibitor binding to confirm on-target effects. The ability of resistant mutants to rescue phenotypes confirms compound specificity .
Temporal controls: Implement time-course experiments, as some ASNA1-dependent phenotypes like Stx5/Stx6 mislocalization develop progressively (normal at E11.5 but severely reduced by E12.5 in knockout models) .
Off-target effect monitoring: Include parallel experiments in ASNA1 knockdown or knockout conditions to distinguish between ASNA1-dependent and -independent effects of compounds.
ASNA1 research has revealed critical roles in organ development, particularly in pancreatic and duodenal tissues. Studies utilizing Asna1Panc−/− mouse models demonstrate that:
Pancreatic development is ASNA1-dependent: Loss of ASNA1 function results in severe dorsal and ventral pancreatic agenesis with reduced branching complexity evident by E15.5 .
ASNA1 prevents apoptosis in developing epithelia: The growth defect in Asna1Panc−/− dorsal pancreatic epithelia coincides with prominent apoptosis at E13.5, particularly in branch tips along the epithelial/mesenchymal border . Quantitative analysis shows:
Golgi integrity depends on ASNA1 function: While Golgi structure and Stx5/Stx6 expression appear normal at E11.5 in knockout models, by E12.5, these proteins show severely reduced immunoreactivity and are virtually absent from the fragmented cis-Golgi and TGN compartments .
These findings suggest potential roles for ASNA1 dysfunction in congenital pancreatic disorders, opening avenues for diagnostic applications of ASNA1 antibodies in developmental pathology.
When investigating ASNA1's role in disease mechanisms, several methodological considerations enhance experimental rigor:
Tissue-specific expression patterns: ASNA1 expression varies across tissues, necessitating validation of antibody performance in each specific context. While the antibody shows reactivity in human brain tissue, HeLa cells, human heart tissue, and mouse brain tissue for Western blotting, optimal dilutions may differ for disease-relevant tissues .
Species cross-reactivity: ASNA1 antibodies have demonstrated reactivity with human, mouse, and rat samples . When studying disease models in other species, cross-reactivity must be experimentally verified before proceeding with extensive analyses.
Subcellular localization studies: For diseases involving protein trafficking defects, subcellular fractionation followed by Western blotting with ASNA1 antibodies can reveal alterations in distribution patterns. This approach should be complemented with immunofluorescence studies examining co-localization with compartment-specific markers .
Protein interaction studies: For investigating disease-relevant ASNA1 interactions, the antibody has been validated for immunoprecipitation in mouse brain tissue, using 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate . This application enables identification of altered protein-protein interactions in disease states.
Maximizing research impact requires integration of antibody-based findings with complementary methodologies:
Multi-omics integration: ASNA1 antibody data can be correlated with:
Transcriptomic data to identify compensatory gene expression changes
Proteomic analyses to map altered interaction networks
Metabolomic studies to connect with cellular stress responses
Functional rescue experiments: When ASNA1 dysfunction is identified, rescue experiments using:
Temporal dynamics: Time-course experiments combining antibody detection with functional readouts can reveal the sequence of events following ASNA1 perturbation, distinguishing primary from secondary effects. For example, in pancreatic development, apoptosis becomes evident at E13.5 while proliferation defects appear later at E15.5 .
In vivo to in vitro translation: Findings from animal models (e.g., Asna1Panc−/− mice) can be validated and mechanistically dissected in cellular systems using approaches like CRISPR-directed diversification and inhibitor studies .
The arsA arsenite transporter ATP-binding homolog 1 is a protein that plays a crucial role in the resistance to arsenicals. This protein is encoded by the ASNA1 gene in humans and is homologous to the bacterial arsA gene. The protein is also known by several other names, including ARSA-I, ARSA1, ASNA-I, GET3, TRC40, and hASNA-I .
The ASNA1 gene is located on chromosome 19 in humans and on chromosome 8 in mice . The gene encodes a protein that is part of a multisubunit oxyanion pump responsible for the transport of arsenite and antimonite ions across cell membranes . The protein has ATPase activity, meaning it can hydrolyze ATP to provide the energy required for the transport process .
In Escherichia coli, the arsA ATPase is the catalytic component of the arsenite transporter system, which provides resistance to arsenicals and antimonials . The human homolog, ASNA1, performs a similar function by acting as an arsenite-stimulated ATPase component of the arsenite transporter . This protein is also involved in the post-translational delivery of tail-anchored (TA) proteins from the cytosol to the endoplasmic reticulum (ER) . It recognizes and selectively binds the transmembrane domain (TMD) of TA proteins in the cytosol and delivers them to the ER for insertion .
The arsA arsenite transporter ATP-binding homolog 1 is essential for cellular resistance to toxic arsenic compounds. By transporting arsenite out of the cell, it helps to mitigate the toxic effects of arsenic exposure . Additionally, its role in the delivery of TA proteins to the ER is crucial for proper cellular function and protein targeting .
Research on the arsA arsenite transporter ATP-binding homolog 1 has provided insights into the mechanisms of arsenic resistance and the broader field of metal ion transport . Understanding this protein’s function can lead to the development of strategies to combat arsenic toxicity and improve bioremediation efforts. Furthermore, the study of its role in TA protein delivery has implications for understanding protein targeting and insertion into the ER .