ACOT9 antibodies are immunological reagents specifically designed to detect and bind to ACOT9 (Acyl-CoA thioesterase 9), a member of the acyl-CoA thioesterase superfamily. These antibodies serve as essential tools for researchers studying lipid metabolism, mitochondrial function, and related metabolic pathways. ACOT9 protein, also known by alternative names such as ACATE2, MT-ACT48 (mitochondrial acyl-CoA thioesterase of 48 kDa), or CGI-16, plays a crucial role in hydrolysis of acyl-CoAs to free fatty acids and coenzyme A (CoA) .
The gene encoding ACOT9 is located on chromosome X, and the expressed ACOT9 protein is primarily localized to the mitochondrion, highlighting its importance in cellular energy metabolism . ACOT9 consists of 406 amino acids and features a C-terminal 80-amino acid domain that is highly conserved from mouse to human, suggesting evolutionary significance and implying that this region may confer the catalytic activity of the enzyme .
ACOT9 protein has a calculated molecular weight ranging from 24 kDa to 49 kDa, though the observed molecular weight in experimental conditions typically falls between 43-50 kDa . The protein exists in four distinct isoforms, contributing to its diverse metabolic functions .
The primary function of ACOT9 is to catalyze the hydrolysis of acyl-CoAs to produce free fatty acids and coenzyme A. Through this catalytic activity, ACOT9 regulates intracellular levels of acyl-CoAs, free fatty acids, and CoA, which are critical components in various metabolic pathways . This regulatory function is essential for maintaining cellular lipid homeostasis and supporting metabolic flexibility during conditions such as fasting or exercise .
Research has demonstrated that ACOT9's hydrolytic activity helps reduce the build-up of acyl-CoA intermediates, which is essential for maintaining cellular energy balance . More specifically, ACOT9 exhibits high activity with saturated long-chain acyl-CoAs but can simultaneously hydrolyze both short-chain and long-chain acyl-CoAs despite markedly different substrate affinities . This dual capability suggests that ACOT9 may provide a novel regulatory link between fatty acid and amino acid metabolism in mitochondria .
ACOT9 antibodies are available in various formats, each with specific characteristics and applications. They can be categorized based on their production methods, host species, and specific properties.
Monoclonal antibodies against ACOT9 are produced from single B-cell clones, ensuring high specificity and consistency between batches. These antibodies target specific epitopes on the ACOT9 protein and are particularly valuable for applications requiring precise antigen recognition.
Several commercial monoclonal ACOT9 antibodies are available, including:
ACOT9 Mouse Monoclonal Antibody (66532-1-PBS) from Proteintech, which is validated in Western Blot, Immunohistochemistry, and Indirect ELISA applications and shows reactivity with human, rat, and mouse samples .
ACOT9 Antibody (O59) from Santa Cruz Biotechnology, an IgG1 κ mouse monoclonal antibody that detects human ACOT9 by Western Blot, Immunoprecipitation, Immunofluorescence, Immunohistochemistry, and ELISA .
ACOT9 Antibody (G-6) from Santa Cruz Biotechnology, an IgG3 κ mouse monoclonal antibody that detects mouse, rat, and human ACOT9 by Western Blot, Immunoprecipitation, Immunofluorescence, and ELISA .
Polyclonal antibodies are produced by immunizing animals with ACOT9 protein or peptide fragments, resulting in antibodies that recognize multiple epitopes on the target protein. This multi-epitope recognition can enhance detection sensitivity but may introduce some variability between batches.
Notable polyclonal ACOT9 antibodies include:
ACOT9 Polyclonal Antibody (PA5-57310) from Thermo Fisher Scientific, generated using an immunogen sequence from the ACOT9 protein .
ACOT9 antibody (15901-1-AP) from Proteintech, a rabbit polyclonal antibody that targets ACOT9 in Western Blot, Immunohistochemistry, and ELISA applications .
ACOT9 Rabbit Polyclonal Antibody (CAB15416) from Assay Genie, produced using a recombinant fusion protein containing a sequence corresponding to amino acids 22-250 of human ACOT9 .
ACOT9 antibodies have been extensively validated for various research applications, enabling scientists to investigate the expression, localization, and function of ACOT9 protein in different biological contexts.
Western blotting is one of the most common applications for ACOT9 antibodies, allowing researchers to detect and quantify ACOT9 protein expression in tissue or cell lysates. Most commercial ACOT9 antibodies are validated for Western blotting, with recommended dilutions typically ranging from 1:500 to 1:2000 .
In Western blot applications, ACOT9 is commonly detected as a band between 43-50 kDa . For example, the Proteintech ACOT9 antibody (15901-1-AP) detects ACOT9 in HEK-293 cells, MCF-7 cells, mouse heart tissue, mouse kidney tissue, and rat kidney tissue .
Immunohistochemistry allows researchers to visualize the distribution and localization of ACOT9 in tissue sections. ACOT9 antibodies validated for IHC have been used to study the protein's expression in various tissues.
For instance, the Proteintech ACOT9 antibody (15901-1-AP) has been validated for IHC in mouse kidney tissue, human kidney tissue, and rat kidney tissue, with recommended dilutions of 1:50 to 1:500 . Similarly, the Abcam Anti-Acyl-CoA Thioesterase 9 antibody (ab252978) has been used to stain paraffin-embedded human bronchus tissue at a 1/1000 dilution .
Immunofluorescence techniques allow for high-resolution imaging of ACOT9 localization within cells. Several ACOT9 antibodies are validated for immunofluorescence applications, enabling researchers to study the subcellular localization of ACOT9.
For example, the Abcam ACOT9 antibody (ab252978) has been used to stain PFA-fixed, Triton X-100 permeabilized U-251MG (Human brain glioma cell line) cells, showing the mitochondrial localization of ACOT9 . The recommended dilution for immunofluorescence applications typically ranges from 1:50 to 1:200 .
In addition to the applications mentioned above, ACOT9 antibodies have been validated for:
Immunoprecipitation (IP): Used to isolate and purify ACOT9 protein from complex mixtures .
Enzyme-Linked Immunosorbent Assay (ELISA): Used for quantitative detection of ACOT9 in solution .
Studies utilizing ACOT9 antibodies have contributed significantly to our understanding of ACOT9's biological function and its role in cellular metabolism.
Research has definitively established that ACOT9 is localized to the mitochondrial matrix. A study by Pougovkina et al. (2019) used validated antibodies specifically targeting Tom20, Tim23, and pyruvate dehydrogenase (PDH) as markers for different mitochondrial compartments, along with an ACOT9 antibody, to determine the precise localization of ACOT9 .
The results showed that in both kidney and heart mitochondria, the 50 kDa band corresponding to ACOT9 disappeared only after adding trypsin plus Triton X-100, similar to the matrix marker PDH. This finding confirmed that ACOT9 is located in the mitochondrial matrix .
Research using ACOT9 antibodies has revealed that ACOT9 plays a significant role in fatty acid metabolism within mitochondria. Studies have shown that ACOT9 has high activity with saturated long-chain acyl-CoAs, suggesting its importance in regulating fatty acid oxidation .
Furthermore, based on similar acyl-CoA chain-length specificities of recombinant ACOT9 and ACOT activity in mouse brown adipose tissue and kidney mitochondria, researchers have concluded that ACOT9 is the major mitochondrial ACOT hydrolyzing saturated C2-C20-CoA in these tissues .
One of the most intriguing findings about ACOT9 is its potential role in connecting fatty acid and amino acid metabolism. Research has shown that ACOT9 can hydrolyze a number of short-chain acyl-CoAs and short-chain methyl-branched CoA esters, which are intermediates or products of amino acid metabolism .
Despite markedly different substrate affinities (Km values), ACOT9 can hydrolyze both short- and long-chain acyl-CoAs simultaneously, indicating that ACOT9 may provide a novel regulatory link between fatty acid and amino acid metabolism in mitochondria .
Additionally, ACOT9 activity is strongly regulated by NADH and CoA, suggesting that the mitochondrial metabolic state regulates the function of ACOT9 . This finding further emphasizes the importance of ACOT9 in coordinating different metabolic pathways within the mitochondria.
The development and validation of ACOT9 antibodies often involve rigorous procedures to ensure specificity and sensitivity. For example, a custom antibody against mouse ACOT9 was developed using a synthetic peptide corresponding to amino acids 225-238 of the mouse ACOT9 protein (with a cysteine added at the N-terminal end for coupling) .
Rabbits were immunized with the peptide conjugated to keyhole limpet hemocyanin, and antibodies were affinity-purified using a column with the peptide conjugated to Epoxy-activated Sepharose 6B . The specificity of the antibody was confirmed by competing out antibody binding to the 48-kDa band with excess recombinant ACOT9 .
Commercial ACOT9 antibodies undergo similar rigorous validation processes. For instance, the specificity of the Acot13 antibody was determined using recombinant Acot13 and liver mitochondria from Acot13 knockout mice, which revealed a single band at approximately 15 kDa in control samples but not in knockout samples .
As our understanding of ACOT9's role in cellular metabolism continues to expand, ACOT9 antibodies will remain essential tools for investigating several promising research areas:
Acyl-CoA thioesterases are a group of enzymes that catalyze the hydrolysis of acyl-CoAs into free fatty acids and coenzyme A (CoASH). This process allows for the regulation of intracellular levels of acyl-CoAs, free fatty acids, and CoASH. This particular antibody is active on long chain acyl-CoAs.
ACOT9 (Acyl-coenzyme A thioesterase 9) is a member of the acyl-CoA thioesterase superfamily, a group of enzymes that catalyze the hydrolysis of acyl-CoAs to free fatty acids and coenzyme A (CoASH). This mitochondrial protein plays a critical role in regulating intracellular levels of acyl-CoAs, free fatty acids, and CoASH. ACOT9 is particularly significant because it can simultaneously hydrolyze both short-chain and long-chain acyl-CoAs despite having markedly different Km values for these substrates . This dual functionality suggests ACOT9 provides a novel regulatory link between fatty acid and amino acid metabolism in mitochondria, making it an important target for metabolic research. Recent studies have also identified ACOT9 as having a strong association with obesity in humans, highlighting its potential importance in metabolism-related disorders .
When selecting an ACOT9 antibody, researchers should carefully evaluate several parameters to ensure experimental success. First, verify species reactivity—commercially available antibodies like 15901-1-AP show reactivity with human, mouse, and rat samples . Second, confirm application compatibility based on your experimental methods; some antibodies are validated for Western blot (1:500-1:1000 dilution), immunohistochemistry (1:50-1:500), ELISA, and immunofluorescence applications . Third, consider the antibody type; most ACOT9 antibodies are rabbit polyclonal, which offers high sensitivity but may show batch-to-batch variation . Fourth, examine the immunogen information—for example, ABClonal's A15416 uses a recombinant fusion protein corresponding to amino acids 22-250 of human ACOT9 . Finally, check if the antibody detects the expected molecular weight (approximately 43-50 kDa for ACOT9) .
ACOT9 primarily localizes to mitochondria but exhibits a dynamic distribution pattern between the inner mitochondrial membrane (IM) and matrix compartments depending on cellular conditions. Under basal conditions, ACOT9 is more abundant in the inner mitochondrial membrane, but it translocates into the matrix upon caloric stress and/or chronic cold exposure . This compartment shifting is functionally significant and appears to involve interaction with the electron transport chain Complex I subunit NDUFS7 . For antibody-based detection, this translocation phenomenon has important implications: immunofluorescence experiments should include mitochondrial markers to confirm colocalization, and subcellular fractionation protocols must carefully separate mitochondrial compartments to accurately track ACOT9 distribution. When performing immunostaining, researchers should consider that fixation methods may differentially preserve the native localization depending on the metabolic state of the cells or tissues being examined.
ACOT9 antibodies have been validated for multiple experimental applications, each requiring specific optimization. For Western blot (WB) applications, researchers have successfully used dilutions ranging from 1:500-1:2000, with recommended starting points of 1:500-1:1000 for Proteintech's 15901-1-AP antibody and 1:1000 for ABClonal's A15416 . Optimal protein loading is approximately 25μg per lane, with detection enhanced using HRP-conjugated secondary antibodies and ECL detection systems. For immunohistochemistry (IHC), dilutions of 1:50-1:500 are recommended, with antigen retrieval in TE buffer (pH 9.0) or alternatively citrate buffer (pH 6.0) . Immunofluorescence/immunocytochemistry (IF/ICC) applications work effectively at dilutions of 1:50-1:200, as demonstrated in A431 cells with Cy3-conjugated secondary antibodies . For all applications, appropriate blocking (e.g., 3% nonfat dry milk in TBST for WB) is essential to minimize background signal. The choice of fixative and permeabilization method should be optimized based on the specific cellular compartment being targeted, particularly considering ACOT9's dynamic mitochondrial localization.
Validating antibody specificity is crucial for ensuring reliable research outcomes. For ACOT9 antibodies, a multi-tiered validation approach is recommended. First, conduct positive and negative control experiments using tissues or cell lines with known ACOT9 expression patterns; validated positive samples include HEK-293 cells, MCF-7 cells, mouse heart tissue, mouse kidney tissue, and rat kidney tissue . Second, verify the molecular weight of detected bands (expected at 43-50 kDa for ACOT9) . Third, consider using ACOT9 knockout or knockdown samples as negative controls; published studies using Acot9-/- mice provide excellent specificity controls . Fourth, when possible, test multiple antibodies targeting different epitopes of ACOT9—for example, compare antibodies raised against different regions of the protein such as the 15901-1-AP (targets a fusion protein) versus A15416 (targets amino acids 22-250) . Finally, consider orthogonal validation methods, such as confirming protein expression with mRNA levels via RT-PCR or demonstrating loss of signal after pre-incubation of the antibody with its immunizing peptide.
For optimal detection of ACOT9 in histological samples, tissue preparation and fixation protocols must preserve both antigenicity and mitochondrial morphology. For formalin-fixed paraffin-embedded (FFPE) sections, 10% neutral buffered formalin fixation for 24-48 hours, followed by standard paraffin embedding, is generally effective. Section thickness of 4-5μm is recommended for IHC applications. Importantly, ACOT9 detection requires optimization of antigen retrieval methods; the primary suggested method is heat-induced epitope retrieval (HIER) using TE buffer at pH 9.0, although citrate buffer at pH 6.0 can serve as an alternative . For frozen sections, fixation with 4% paraformaldehyde for 10-15 minutes followed by permeabilization with 0.2% Triton X-100 typically provides good results. When studying the submitochondrial localization of ACOT9 in research investigating its translocation between inner membrane and matrix compartments, electron microscopy with immunogold labeling may be necessary, requiring specialized fixation with glutaraldehyde/paraformaldehyde mixtures followed by careful dehydration and embedding in resin.
ACOT9 exhibits a complex substrate specificity profile that requires careful consideration when designing activity assays. Research has demonstrated that ACOT9 can hydrolyze both short-chain and long-chain acyl-CoAs simultaneously despite having significantly different Km values for these substrates . When designing ACOT9 activity assays, researchers should consider including multiple substrate types, including short-chain acyl-CoAs (C2-C6), medium-chain acyl-CoAs (C8-C12), long-chain acyl-CoAs (C14-C20), and branched-chain CoA esters derived from amino acid metabolism. For acyl-CoA thioesterase activity assays, HPLC-based detection methods using UV detection at 260nm have been successfully employed, using an appropriate internal standard such as 3-hydroxy-3-methylglutaryl-CoA (which is not a substrate for ACOT9) . The reaction should be conducted in a suitable buffer system, with reactions terminated by acidification to pH 4 to inhibit ACOT9 activity. Additionally, researchers should be aware that ACOT9 activity is strongly regulated by NADH and CoA, so the concentration of these metabolites in the reaction mixture will significantly impact measured activity levels .
Investigating ACOT9 submitochondrial translocation requires specialized approaches to distinguish between inner membrane and matrix localization. Based on recent findings that ACOT9 translocates from the inner membrane to the matrix upon caloric stress and chronic cold exposure , researchers can employ several complementary methods. First, subcellular fractionation with differential centrifugation can be used to isolate mitochondria, followed by further separation of mitochondrial compartments using digitonin treatment or osmotic shock techniques to separate outer membrane, inner membrane, and matrix fractions. Western blot analysis can then quantify ACOT9 distribution across these fractions, using compartment-specific markers (e.g., TOM20 for outer membrane, TIM23 for inner membrane, and matrix enzymes like citrate synthase) as controls. Second, super-resolution microscopy with co-localization studies using compartment-specific markers can visualize ACOT9 translocation in intact cells. Third, proximity labeling approaches like BioID or APEX2 fused to compartment-specific anchors can track ACOT9 movement in living cells. Finally, researchers should consider monitoring the association between ACOT9 and its binding partner NDUFS7 (a Complex I subunit), as this interaction may regulate the translocation process .
To investigate ACOT9's proposed role as a link between fatty acid and amino acid metabolism, researchers can implement multi-faceted experimental approaches. First, enzyme kinetic studies using recombinant ACOT9 and various substrates including fatty acyl-CoAs and CoA esters derived from amino acid catabolism can establish substrate preferences and potential competitive relationships. HPLC-based assays have been successfully used for this purpose, measuring the hydrolysis of substrates like C12-CoA and isobutyryl-CoA using 3-hydroxy-3-methylglutaryl-CoA as an internal standard . Second, metabolomic profiling of cells or tissues with ACOT9 overexpression or knockout (Acot9-/-) can reveal changes in metabolites from both pathways. Studies using Acot9-/- mice have already demonstrated effects on energy expenditure and adiposity . Third, stable isotope tracing experiments using labeled fatty acids or amino acids can track metabolic flux alterations when ACOT9 levels are manipulated. Fourth, protein interaction studies can identify ACOT9 binding partners involved in either pathway; NDUFS7 has already been identified as an interaction partner . Finally, tissue-specific knockout models, such as the BAT-specific knockout (Acot9B-KO) that affects body weight and fat mass, can help delineate tissue-specific functions .
When working with ACOT9 antibodies, researchers may encounter several technical challenges. One common issue is inconsistent detection of the expected 43-50 kDa band in Western blot applications. This may result from sample preparation conditions that disrupt mitochondrial integrity or protein degradation. To address this, use fresh samples, include protease inhibitors in all buffers, maintain samples at 4°C throughout processing, and optimize lysis conditions (e.g., RIPA buffer with 0.1% SDS for complete extraction). Another challenge is high background in immunostaining applications, which can be mitigated by optimizing blocking conditions (3% BSA or 5-10% normal serum from the same species as the secondary antibody), extending blocking time to 1-2 hours, and using more stringent washing protocols (e.g., 5x5 minute washes with 0.1% Tween-20 in PBS). For tissue-specific variations in signal intensity, researchers should adjust antibody concentrations based on the tissue type; kidney tissue typically shows strong ACOT9 expression and can serve as a positive control . If multiple bands appear in Western blots, this may reflect ACOT9 isoforms or post-translational modifications; confirmation with mass spectrometry or isoform-specific primers can help clarify the identity of these bands.
Integrating ACOT9 antibody-based detection with functional studies requires a coordinated experimental approach. Researchers can correlate ACOT9 protein levels or subcellular localization with functional readouts of mitochondrial metabolism using the following strategies: First, perform parallel analyses of ACOT9 expression (via Western blot) and mitochondrial respiration (using Seahorse XF analyzers or Clark-type oxygen electrodes) in the same samples under various conditions, such as nutrient deprivation or cold exposure known to trigger ACOT9 translocation . Second, combine immunofluorescence imaging of ACOT9 with live-cell mitochondrial functional probes (e.g., TMRM for membrane potential, MitoTracker for mitochondrial mass, or genetically-encoded sensors for ATP, ROS, or Ca2+) to correlate localization with functional parameters at the single-cell level. Third, use ACOT9 antibodies for immunoprecipitation followed by activity assays to link protein interaction partners with functional outcomes. Fourth, in animal models, correlate tissue-specific ACOT9 expression patterns with physiological parameters; for example, in Acot9B-KO mice, reduced BAT ACOT9 expression correlates with 13% lower body weight and 43% reduction in fat mass after HFD feeding compared to controls .
When studying ACOT9 in disease models or patient samples, several important considerations should guide experimental design and interpretation. First, ACOT9 expression and localization may be altered in disease states, particularly those involving metabolic dysfunction; hence, careful selection of control samples and standardization of collection and processing protocols is essential. Second, given ACOT9's strong association with obesity in humans , researchers should comprehensively document metabolic parameters of study subjects or animal models, including body mass index, fat distribution, insulin sensitivity, and feeding status. Third, when analyzing patient samples, consider potential effects of medications, particularly those affecting mitochondrial function or metabolism (e.g., metformin, thiazolidinediones, statins) on ACOT9 expression or function. Fourth, for tissue biopsies, the timing of sample collection relative to fasting/feeding cycles is critical, as ACOT9 translocation is responsive to caloric stress . Finally, researchers should consider genetic variation in ACOT9 when studying diverse human populations; genetic analysis or sequencing may be necessary to identify functional variants that could affect antibody epitopes or protein function. For mouse models of human disease, note that while mouse and human ACOT9 share high homology, species-specific differences in regulation or interacting partners may exist.
Advanced imaging technologies offer powerful approaches for investigating ACOT9's dynamic subcellular localization and function. Super-resolution microscopy techniques such as structured illumination microscopy (SIM), stimulated emission depletion (STED), or single-molecule localization methods (PALM/STORM) can resolve ACOT9 distribution within mitochondrial subcompartments at resolutions below 100 nm. These techniques can visualize the translocation of ACOT9 from the inner membrane to the matrix under stress conditions with unprecedented clarity . Live-cell imaging combined with ACOT9 fusion proteins (e.g., ACOT9-GFP) enables real-time tracking of translocation events in response to metabolic perturbations, though researchers must verify that the tag doesn't interfere with localization or function. Correlative light and electron microscopy (CLEM) can bridge the resolution gap between fluorescence microscopy and ultrastructural analysis, providing context for ACOT9 localization within the complex mitochondrial architecture. Förster resonance energy transfer (FRET) or bioluminescence resonance energy transfer (BRET) approaches can investigate ACOT9's interaction with identified binding partners like NDUFS7 in living cells. Finally, emerging proximity labeling techniques such as APEX2 or TurboID fused to ACOT9 can identify novel interaction partners in different submitochondrial compartments under various physiological conditions.
Genetic approaches provide powerful complementary strategies to antibody-based ACOT9 studies. CRISPR/Cas9 genome editing can generate precise knockout models in cell lines and animals, as demonstrated in the generation of Acot9-/- and tissue-specific knockout (e.g., Acot9B-KO) mouse models that revealed ACOT9's role in energy expenditure and adiposity regulation . For temporal control of ACOT9 expression, inducible knockdown or knockout systems (e.g., Tet-On/Off or tamoxifen-inducible Cre-loxP) allow researchers to distinguish between developmental and acute effects of ACOT9 loss. Point mutations targeting specific functional domains can dissect the structure-function relationships of ACOT9, particularly regarding its dual substrate specificity for short- and long-chain acyl-CoAs . Rescue experiments reintroducing wild-type or mutant ACOT9 into knockout backgrounds can confirm phenotype specificity and map functional domains. RNA-sequencing of tissues from Acot9-deficient models can reveal transcriptional networks affected by ACOT9 loss, while ribosome profiling might uncover translational effects. Notably, these genetic approaches should be designed with consideration of ACOT9's multiple isoforms and potential compensation by other acyl-CoA thioesterase family members.
Metabolomic approaches offer unique insights into ACOT9 function by directly measuring its impact on cellular metabolism. Targeted metabolomics focusing on acyl-CoA species, free fatty acids, and amino acid metabolites can quantify ACOT9's substrate and product pools in different tissues and under various physiological conditions. This is particularly relevant given ACOT9's ability to hydrolyze both fatty acyl-CoAs and CoA esters derived from amino acid metabolism, potentially providing a metabolic link between these pathways . Stable isotope tracing experiments using 13C-labeled fatty acids or amino acids can track metabolic flux alterations when ACOT9 levels are manipulated, revealing how ACOT9 activity redirects carbon flow through different pathways. Flux balance analysis integrating metabolomic data with computational modeling can predict system-wide effects of ACOT9 perturbation. Spatial metabolomics techniques such as MALDI imaging mass spectrometry can map metabolite distribution in tissues from wild-type versus Acot9-/- or Acot9B-KO mice, potentially correlating with ACOT9 expression patterns identified by immunohistochemistry . Finally, integration of metabolomic data with transcriptomic and proteomic datasets from the same experimental systems can provide a comprehensive multi-omics view of how ACOT9 influences cellular metabolism beyond what can be determined from antibody-based detection alone.