ACS10 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
ACS10 antibody; At1g62960 antibody; F16P17.11Probable aminotransferase ACS10 antibody; EC 2.6.1.- antibody
Target Names
ACS10
Uniprot No.

Target Background

Function
This antibody targets a probable aminotransferase. Enzyme assays indicate a lack of 1-aminocyclopropane-1-carboxylate synthase (ACS) activity, suggesting it is not involved in ethylene biosynthesis.
Database Links

KEGG: ath:AT1G62960

STRING: 3702.AT1G62960.1

UniGene: At.14857

Protein Families
Class-I pyridoxal-phosphate-dependent aminotransferase family
Tissue Specificity
Expressed in roots.

Q&A

What is ACS10 and why is it significant in biomedical research?

ACS10 (acyl-CoA synthetase 10) is an enzyme with critical functions in cellular metabolism across various organisms. In human research, ACS10 has gained significance through the development of the ACS10 score, a pharmacogenomic tool comprising 10 SNPs relevant to cytarabine (ara-C) metabolism in AML treatment . In parasitology, PfACS10 (Plasmodium falciparum acyl-CoA synthetase 10) has been identified as an essential enzyme for parasite lipid metabolism and survival .

To study ACS10, researchers develop antibodies that specifically target this protein to investigate its expression levels, localization, and interactions with other cellular components. These antibodies serve as critical tools in understanding ACS10's role in normal physiology and disease pathology, enabling researchers to track the protein in different experimental conditions and tissue samples.

How do ACS10 antibodies compare with other research tools for studying ACS10 function?

ACS10 antibodies offer unique advantages over other research methods such as genetic manipulation or small molecule inhibitors. While CRISPR-Cas9 or RNAi techniques provide insights into gene function through knockout or knockdown approaches, antibodies enable protein-level detection without altering the underlying genetic code.

Compared to small molecule inhibitors like MMV665924, MMV019719, and MMV897615 that target PfACS10 activity , antibodies allow for:

  • Visualization of protein localization through immunofluorescence microscopy

  • Quantification of protein expression through Western blotting

  • Isolation of protein complexes through immunoprecipitation

  • Validation of target engagement in thermal proteome profiling experiments

These advantages make antibodies complementary to genetic and chemical approaches in comprehensive ACS10 research programs, especially when studying protein-protein interactions that may be critical to understanding ACS10's role in disease processes.

What are the current challenges in generating specific antibodies against ACS10?

Generating highly specific antibodies against ACS10 presents several technical challenges:

  • Homology with other ACS family members can lead to cross-reactivity, particularly with closely related enzymes like ACS11, which has been implicated in resistance mechanisms

  • Post-translational modifications of ACS10 in different cellular contexts might affect epitope accessibility

  • Species-specific variations in ACS10 structure require careful design for cross-species studies

  • Limited availability of purified ACS10 protein for immunization and validation processes

Researchers can address these challenges through:

  • Selecting unique peptide sequences as immunogens to reduce cross-reactivity

  • Validating antibody specificity using knockout/knockdown models

  • Developing recombinant antibody technologies for enhanced reproducibility, similar to approaches used for other immunotherapy targets

  • Implementing rigorous validation protocols to ensure antibody performance across different experimental conditions

How do ACS10 antibodies contribute to understanding pharmacogenomic mechanisms in AML treatment?

ACS10 antibodies provide critical tools for investigating the molecular mechanisms underlying the ACS10 score's predictive value in AML treatment. The pharmacogenomic ACS10 score, which incorporates 10 SNPs related to ara-C metabolism, has demonstrated significant associations with clinical outcomes in pediatric AML patients .

Methodologically, researchers can use ACS10 antibodies to:

  • Compare protein expression levels between patients with high versus low ACS10 scores

  • Investigate correlations between SNP variants and ACS10 protein function through activity assays

  • Examine cellular localization patterns in patient-derived samples

  • Study protein interactions that might be affected by genetic variations

These applications help translate genetic associations into mechanistic understandings, potentially revealing how SNPs influence protein expression, activity, or interactions in ways that affect treatment response.

What experimental protocols are most effective for studying ACS10 protein expression in patient samples using antibodies?

When examining ACS10 protein expression in patient samples, researchers should consider these methodological approaches:

  • Sample preparation:

    • Fresh frozen tissue provides optimal protein preservation for antibody studies

    • FFPE (formalin-fixed paraffin-embedded) samples require antigen retrieval optimization

    • Peripheral blood samples should undergo controlled isolation procedures to minimize degradation

  • Detection protocols:

    • Immunohistochemistry with careful titration of antibody concentrations

    • Multiplex immunofluorescence to correlate ACS10 expression with cell type markers

    • Flow cytometry for quantitative single-cell analysis in blood samples

  • Validation approaches:

    • Include positive controls with known ACS10 expression

    • Use multiple antibodies targeting different epitopes when possible

    • Correlate protein expression with mRNA levels from the same samples

When analyzing results, researchers should stratify patients by ACS10 score (≤0 as low, >0 as high) to investigate whether protein expression patterns correlate with the genetic score and clinical outcomes .

How can ACS10 antibodies be utilized to investigate racial disparities in AML treatment outcomes?

Research has revealed significant racial disparities in ACS10 scores, with approximately 70% of Black patients having low-ACS10 scores compared to approximately 30% of White patients . These differences appear to be driven by three key SNPs with varying allele frequencies across racial groups:

SNPGeneImpactWhite Allele FrequencyBlack Allele Frequency
rs4643786DCKDetrimental0.0380.48
rs1044457CMPK1Beneficial0.50.11
rs17343066SLC28A3Beneficial0.530.15

To investigate these disparities, researchers can use ACS10 antibodies to:

  • Compare protein expression and function across racial groups in healthy controls

  • Examine whether SNP-associated changes in expression correlate with antibody staining patterns

  • Develop tissue microarrays with demographically diverse samples to assess protein expression patterns systematically

  • Investigate whether augmented therapy approaches (HDAC or ADE+GO) differentially affect ACS10 protein levels or activity across racial groups

These approaches may help translate genetic observations into actionable insights for addressing treatment disparities, potentially informing pharmacogenomic-guided treatment decisions similar to established approaches for TPMT and NUDT15 in acute lymphoblastic leukemia .

What are the optimal conditions for using ACS10 antibodies in thermal proteome profiling experiments?

Thermal proteome profiling (TPP) has been successfully used to validate ACS10 as a target of antimalarial compounds . When designing TPP experiments with ACS10 antibodies, researchers should consider:

  • Sample preparation:

    • Cell lysis conditions should preserve ACS10 native structure (mild detergents recommended)

    • Temperature gradient typically from 37°C to 67°C in 3°C increments

    • Compound concentrations should include EC50 value and 5-10x EC50

  • Antibody selection considerations:

    • Choose antibodies recognizing epitopes unlikely to be affected by thermal denaturation

    • Validate antibody performance at different temperatures prior to full experiments

    • Consider using multiple antibodies targeting different epitopes to confirm results

  • Data analysis approach:

    • Plot relative ACS10 signal intensity versus temperature to generate melting curves

    • Calculate melting temperature (Tm) shifts between compound-treated and control samples

    • Normalize to known housekeeping proteins unaffected by the compounds

These experiments can definitively establish whether candidate compounds directly engage with ACS10 protein, as demonstrated with the antimalarial compounds MMV665924, MMV019719, and MMV897615 .

How can ACS10 antibodies be integrated with genetic approaches to validate target engagement?

A multifaceted approach combining antibodies with genetic manipulation provides the strongest evidence for ACS10 target validation:

  • Complementary methodologies:

    • Conditional knockdown models (as used for PfACS10) to demonstrate essentiality

    • Allelic replacement of wild-type with mutant forms harboring resistance mutations

    • CRISPR interference to modulate expression levels

  • Antibody applications in genetic models:

    • Confirm knockdown/knockout efficiency at protein level

    • Visualize subcellular redistribution in response to genetic manipulation

    • Assess protein stability changes from introduced mutations (e.g., M300I, A268D/V, F427L in PfACS10)

  • Target engagement workflow:

    • Generate resistant parasite lines through compound pressure

    • Identify mutations through whole-genome sequencing

    • Use antibodies to confirm altered protein levels or stability

    • Perform thermal proteome profiling to validate direct binding changes

This integrated approach provided compelling evidence for PfACS10 as the target of novel antimalarial compounds, demonstrating how antibodies complement genetic methods in target validation .

What are the methodological considerations for studying ACS10 interactions with lipid metabolism?

Inhibition of PfACS10 has been shown to reduce triacylglycerols and cause buildup of lipid precursors . To investigate similar mechanisms in other systems, researchers should consider:

  • Lipid profiling methodologies:

    • Lipidomics using LC-MS/MS for comprehensive lipid species identification

    • Thin-layer chromatography for rapid screening of major lipid classes

    • Fluorescent lipid analogs to track metabolic flux in live cells

  • Antibody-based approaches:

    • Co-immunoprecipitation to identify protein interaction partners in lipid metabolism

    • Proximity ligation assays to visualize ACS10 interactions with metabolic enzymes

    • Immunofluorescence co-localization with lipid droplet markers

  • Experimental design recommendations:

    • Include appropriate time points to capture dynamic lipid metabolism changes

    • Compare effects of antibody-mediated neutralization versus small molecule inhibition

    • Correlate lipid profile changes with functional outcomes (e.g., parasite survival)

These approaches can illuminate ACS10's role in lipid metabolism across different biological systems, building on the findings from Plasmodium research while extending to other contexts such as cancer cell metabolism.

How can researchers address non-specific binding when using ACS10 antibodies in complex samples?

Non-specific binding is a common challenge when using antibodies in complex biological samples. For ACS10 antibodies, consider these specialized approaches:

  • Optimization strategies:

    • Titrate antibody concentrations across a wider range than standard protocols

    • Test multiple blocking agents (BSA, serum, commercial blockers) for optimal signal-to-noise ratio

    • Implement additional washing steps with increased stringency for high-background samples

  • Validation controls:

    • Use ACS10 knockout/knockdown samples as negative controls

    • Include competitive blocking with immunizing peptides

    • Perform parallel staining with multiple ACS10 antibodies targeting different epitopes

  • Advanced techniques for complex samples:

    • Pre-absorb antibodies against tissue/cell lysates from knockout models

    • Employ antigen retrieval optimization matrices for FFPE samples

    • Consider recombinant engineered antibody formats that can reduce non-specific binding

These approaches are particularly important when studying ACS10 in diverse patient samples or across species barriers, where epitope conservation and sample preparation variables can significantly impact specificity.

What strategies can resolve contradictory results between ACS10 protein expression and functional outcomes?

Researchers may encounter situations where ACS10 antibody staining patterns don't align with expected functional outcomes based on genetic data or treatment responses. To resolve such contradictions:

  • Technical considerations:

    • Verify antibody lot consistency and specificity using reference standards

    • Assess whether post-translational modifications might affect epitope recognition

    • Consider whether sample processing might selectively impact certain protein conformations

  • Biological explanations:

    • Investigate potential compensatory mechanisms (e.g., upregulation of ACS11)

    • Examine whether protein expression changes correlate with activity levels

    • Consider heterogeneity within sample populations that might mask correlations

  • Integrated analysis approaches:

    • Correlate genomic data (ACS10 score), protein expression, and clinical outcomes in the same samples

    • Develop multivariate models incorporating additional biomarkers

    • Perform longitudinal studies to capture dynamic changes in expression and function

This integrated approach is particularly relevant when studying ACS10 in AML patients, where the relationship between the ACS10 score and treatment outcomes varies by therapy type .

How should researchers design ACS10 antibody experiments to investigate interplay between ACS10 and ACS11?

The interplay between ACS10 and ACS11 appears significant, with mutations in ACS11 (F387V, D648Y, and E668K) potentially mediating resistance mechanisms through decreased protein stability . To investigate this relationship:

  • Experimental design considerations:

    • Develop co-staining protocols with antibodies against both proteins

    • Establish dual knockdown/knockout models to assess compensatory mechanisms

    • Use proximity-based assays (FRET, PLA) to detect potential physical interactions

  • Sample preparation recommendations:

    • Fractionate cellular components to determine co-localization in specific compartments

    • Preserve protein-protein interactions through mild lysis conditions

    • Consider crosslinking approaches to stabilize transient interactions

  • Data analysis framework:

    • Quantify relative expression levels of ACS10 and ACS11 across experimental conditions

    • Assess correlation patterns between expression levels and functional outcomes

    • Develop mathematical models of potential compensatory mechanisms

This approach could clarify whether ACS11 serves primarily as a resistance mechanism or has complementary functions in normal cellular metabolism, insights that would be valuable for both AML treatment and antimalarial drug development .

How might ACS10 antibodies contribute to developing personalized medicine approaches for AML?

ACS10 antibodies could become valuable tools in translating the ACS10 score into clinical practice through:

  • Development of diagnostic assays:

    • Immunohistochemistry-based scoring systems correlating protein expression with genetic score

    • Flow cytometry panels for rapid assessment of ACS10 status in patient samples

    • Point-of-care tests to complement genomic testing for treatment decision-making

  • Treatment monitoring applications:

    • Serial assessment of ACS10 expression during therapy to detect adaptive changes

    • Correlation of protein levels with minimal residual disease (MRD) status

    • Identification of patients who might benefit from augmented therapy approaches (HDAC or ADE+GO)

  • Integration with electronic health records:

    • Development of algorithms incorporating both genetic and protein expression data

    • Implementation of web-based tools for calculating comprehensive risk scores

    • Standardization of reporting to facilitate multicenter studies and data sharing

These approaches could accelerate clinical translation similar to established pharmacogenomic applications such as TPMT and NUDT15 in acute lymphoblastic leukemia .

What methodological advancements are needed to study ACS10 in combination with immune checkpoint molecules?

Investigating potential interactions between ACS10 and immune checkpoint molecules could open new therapeutic avenues. Researchers should consider:

  • Antibody engineering approaches:

    • Development of bispecific antibodies targeting both ACS10 and checkpoint molecules

    • Creation of antibody panels for multiplexed immunophenotyping

    • Engineering species cross-reactive antibodies for translational studies

  • Experimental systems:

    • Co-culture models with both malignant cells and immune components

    • Patient-derived xenografts with humanized immune systems

    • Ex vivo analysis of patient samples before and after immunotherapy

  • Analytical considerations:

    • Multiplex imaging to visualize ACS10 expression in the tumor microenvironment

    • Single-cell analysis to correlate ACS10 with checkpoint molecule expression

    • Computational approaches to identify potential regulatory relationships

These methodologies could reveal whether ACS10 status influences response to immunotherapies, potentially identifying new combination approaches for patients with low ACS10 scores who currently show poorer outcomes with standard treatments .

How can ACS10 antibody research contribute to addressing racial disparities in cancer treatment?

The observation that approximately 70% of Black patients versus approximately 30% of White patients have low-ACS10 scores suggests this pathway may contribute to documented racial disparities in AML outcomes . To address this:

  • Research priorities:

    • Develop antibody-based assays to rapidly identify patients with low ACS10 protein expression

    • Investigate whether protein levels correlate with genetic scores across racial groups

    • Determine whether augmented therapy approaches equally benefit all populations

  • Methodological considerations:

    • Ensure validation cohorts include diverse racial and ethnic populations

    • Develop standardized protocols that perform consistently across different demographic groups

    • Create reference standards for antibody-based assays that account for population variations

  • Translation to practice:

    • Design clinical trials stratified by both ACS10 score and self-reported race/ethnicity

    • Develop risk calculators incorporating both genetic and antibody-based data

    • Implement preemptive genotyping and protein expression analysis in clinical settings

This targeted approach could help reduce outcome disparities by ensuring that treatment decisions account for both genetic and protein-level variations that affect drug response across different populations .

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.