ABHD14B antibodies are typically rabbit-derived polyclonal reagents validated for use in techniques like Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF/ICC), and ELISA. Key features include:
| Property | Details |
|---|---|
| Target | ABHD14B (UniProt: Q96IU4) |
| Molecular Weight | ~22–25 kDa (observed vs. 22 kDa calculated) |
| Immunogen | Recombinant full-length protein or synthesized peptides (e.g., residues 161–210) |
| Host Species | Rabbit |
| Reactivity | Human, mouse, rat |
ABHD14B antibodies have been rigorously tested across multiple platforms:
Western Blot (WB): Detects endogenous ABHD14B in lysates (e.g., mouse liver, spleen) at dilutions of 1:500–1:2400 .
Immunohistochemistry (IHC): Localizes ABHD14B in formalin-fixed tissues (e.g., human prostate hyperplasia, spleen) using antigen retrieval (TE buffer pH 9.0) and dilutions of 1:20–1:200 .
Immunofluorescence (IF/ICC): Visualizes ABHD14B in cell lines like HepG2 at 1:10–1:100 dilutions .
Use fresh tissue lysates for WB to avoid protein degradation.
For IHC, citrate buffer (pH 6.0) serves as an alternative antigen retrieval method .
ABHD14B antibodies have been pivotal in elucidating the enzyme's biological roles:
Metabolic Regulation: Knockdown studies in HEK293T cells revealed ABHD14B's critical role in glycolysis and the TCA cycle, with depletion leading to reduced cellular lactate and pyruvate levels .
Tissue Distribution: ABHD14B is highly expressed in metabolically active tissues (e.g., liver, kidneys), as confirmed by IHC and WB .
Enzymatic Function: Antibody-based validation confirmed ABHD14B's lysine deacetylase activity, which generates acetyl-CoA from acetylated lysine residues .
Specificity: Antibodies show no cross-reactivity with unrelated hydrolases, confirmed by siRNA knockdown and negative controls .
Functional Studies: Used to validate ABHD14B's interaction with transcriptional regulators (e.g., TFIID) and its impact on acetyl-CoA levels .
Species Cross-Reactivity: Consistent detection in human, mouse, and rat models .
ABHD14B (Abhydrolase Domain Containing 14B) is an enzyme belonging to the metabolic serine hydrolase family. It has been functionally annotated as a lysine deacetylase (KDAC) that transfers an acetyl group from post-translationally modified protein lysine residues to coenzyme A (CoA), generating acetyl-CoA and regenerating the free amine of protein lysine residues .
ABHD14B was initially identified as CCG1/TAF II250-interacting factor B (CIB) through a yeast two-hybrid screen seeking interacting partners of the histone acetyltransferase (HAT) domain of the largest TFIID transcription factor subunit . Recent multi-omics analyses reveal that ABHD14B plays a significant role in regulating glucose metabolism, with its loss resulting in altered glucose metabolism pathways .
ABHD14B shows restricted tissue expression patterns. Using selective antibodies against mammalian ABHD14B, tissue distribution surveys in mice have revealed that ABHD14B is predominantly expressed in metabolically active tissues, particularly the liver and kidneys . Additional immunohistochemistry data indicates ABHD14B expression in:
| Tissue Type | Detection Level |
|---|---|
| Liver | High expression |
| Kidney | High expression |
| Small intestine | Detectable |
| Prostate | Detectable |
| Spleen | Detectable |
This restricted expression pattern in metabolically active tissues suggests ABHD14B may play an important role in regulating metabolism and cellular energetics .
ABHD14B possesses the canonical ABHD fold with an invariant catalytic triad (Ser-His-Asp). The crystal structure of human ABHD14B (PDB: 1IMJ) was determined over a decade ago, revealing that it contains the nucleophilic serine residue (S111) as part of a non-canonical SxxS motif . Based on its protein sequence, ABHD14B is categorized as an outlying member of the metabolic serine hydrolase family, which comprises approximately 1-2% of the total proteome in mammals .
ABHD14B represents a novel class of lysine deacetylases distinct from the well-studied sirtuins and histone deacetylase (HDAC) enzymes . While all these enzyme families can deacetylate protein lysine residues, they employ different mechanisms:
| Deacetylase Family | Cofactor Requirement | Mechanism |
|---|---|---|
| ABHD14B | Requires CoA | Transfers acetyl group from acetylated lysine to CoA, generating acetyl-CoA |
| Sirtuins | Requires NAD+ | Cleaves NAD+ during deacetylation, generating O-acetyl-ADP-ribose |
| HDACs (Class I, II, IV) | Requires Zn2+ | Hydrolytic mechanism using water as nucleophile |
This mechanistic distinction makes ABHD14B particularly interesting for researchers studying metabolic regulation, as its activity directly contributes to acetyl-CoA production, a central metabolic intermediate .
Transcriptomics and metabolomics analyses in ABHD14B knockdown mammalian cells (HEK293T) reveal that ABHD14B significantly influences glucose metabolism. When ABHD14B is depleted:
Cellular PKAc levels increase
Cellular acetyl-CoA concentrations decrease
Glucose metabolism is altered, with effects on both glycolysis and TCA cycle
Lactate production is affected, with depleted cellular lactate levels and increased secreted lactate (approximately 2-fold)
Network analysis of differentially expressed genes (DEGs) showed that cellular or primary metabolic processes (particularly glycolysis and the citric acid cycle) were the most overrepresented pathway annotations affected by ABHD14B knockdown .
Given the current lack of specific pharmacological inhibitors and genetic animal models for ABHD14B, researchers have employed several alternative approaches:
In vivo nonviral transfection: To knockdown hepatic ABHD14B in mice using validated plasmids (e.g., KD_2 and KD_3 plasmids that have demonstrated >95% knockdown efficiency in mammalian cells)
Multi-omics approach: Combining transcriptomics and metabolomics analyses to comprehensively understand the metabolic consequences of ABHD14B depletion
Tissue-specific functional studies: Given ABHD14B's restricted expression in metabolically active tissues, targeting liver-specific functions during different metabolic states (e.g., fasting conditions)
Biochemical assays: Using recombinantly purified human ABHD14B for in vitro deacetylation assays with CoA as substrate
These approaches have revealed that disruption of hepatic ABHD14B, especially during fasting, disturbs homeostatic systemic glucose metabolism and significantly alters organismal energetic status .
Based on validated commercial antibodies, the recommended dilutions vary by application:
For optimal results, it is recommended to titrate the antibody for each testing system, as the optimal dilution may be sample-dependent .
For optimal detection of ABHD14B in tissue samples by immunohistochemistry:
Antigen retrieval recommendations:
Tissue types with validated positive detection:
Fixation considerations: Standard formalin fixation followed by paraffin embedding (FFPE) is compatible with most commercially available ABHD14B antibodies .
To ensure antibody specificity and minimize false results:
Genetic validation:
Tissue distribution validation:
Western blot molecular weight verification:
Positive and negative controls:
Based on research literature, the following cell lines have been successfully used for ABHD14B studies:
For functional studies, HEK293T cells have been effectively used with plasmid-based knockdown approaches (particularly plasmids KD_2 and KD_3) to study the metabolic consequences of ABHD14B depletion .
For robust ABHD14B knockdown experiments, include:
Non-targeting (NT) control plasmid: To control for non-specific effects of the transfection procedure
Untreated "wild-type" cells: To control for potential "off-target" effects of the NT plasmid
Multiple distinct knockdown constructs: Using two or more different knockdown plasmids (e.g., KD_2 and KD_3) targeting different regions of ABHD14B mRNA helps validate that observed phenotypes are due to ABHD14B depletion rather than off-target effects
qPCR validation: Confirm knockdown efficiency at the mRNA level
Western blot validation: Verify protein depletion using a validated ABHD14B antibody
Rescue experiment: Where possible, include a rescue condition with ABHD14B re-expression to confirm specificity of observed phenotypes
To specifically attribute deacetylation activity to ABHD14B rather than other cellular deacetylases:
Cofactor dependency analysis:
Inhibitor profiling:
Substrate specificity analysis:
Enzymatic assay conditions:
For successful detection of ABHD14B in tissue samples:
Tissue selection: Prioritize liver and kidney tissues where ABHD14B is highly expressed
Antigen retrieval optimization:
Antibody dilution titration: Start with manufacturer's recommended range (typically 1:20-1:200) and optimize for your specific tissue samples
Detection system selection: Both chromogenic and fluorescent detection systems have been successfully used with ABHD14B antibodies
Controls:
Include positive control tissues (liver, kidney, small intestine)
Include negative control tissues where ABHD14B is not expressed
Include technical negative controls (omitting primary antibody)
Background reduction: If background is problematic, consider:
Additional blocking steps
Higher dilution of primary antibody
Shorter incubation time
Alternative detection systems
Given ABHD14B's role in glucose metabolism and its restricted expression in metabolically active tissues, several research directions are promising:
Metabolic syndrome and diabetes: Investigate if ABHD14B dysregulation contributes to impaired glucose homeostasis
Liver metabolism: Study ABHD14B's role in hepatic glucose metabolism during fasting/feeding cycles
Acetyl-CoA regulation: Explore how ABHD14B contributes to the cellular acetyl-CoA pool, which affects various metabolic pathways
Transcriptional regulation: Investigate ABHD14B's potential role in regulating transcription of metabolic genes through its interaction with transcription factors
Development of specific inhibitors: Design and test small molecule inhibitors of ABHD14B to assess therapeutic potential in metabolic diseases
Research approaches should integrate transcriptomics, metabolomics, and targeted functional studies in both cellular and animal models to fully understand ABHD14B's role in metabolic regulation .
While not explicitly detailed in the available search results, several emerging techniques would be valuable for ABHD14B research:
Proximity labeling approaches: BioID or APEX2 fusion proteins to identify physiological interaction partners of ABHD14B
Activity-based protein profiling (ABPP): To identify specific substrates and develop selective inhibitors for ABHD14B
CRISPR-based genetic models: Development of ABHD14B knockout mice or cell lines for definitive functional studies
Chemical genetics: Using engineered ABHD14B variants that can be specifically inhibited by bulky inhibitors
Proteomic identification of acetylation sites: Global acetylome analysis after ABHD14B manipulation to identify its physiological substrates
These approaches would address current gaps in our understanding of ABHD14B biology and potentially reveal new therapeutic targets for metabolic diseases.