ACAT2 antibodies are pivotal in studying lipid metabolism and disease mechanisms:
Hepatic Localization: ACAT2 antibodies confirmed its exclusive expression in human hepatocytes (via IHC) and its role in esterifying cholesterol for VLDL/LDL secretion .
Enterocyte Studies: Used to validate ACAT2’s function in dietary cholesterol absorption and chylomicron assembly .
Gastric and Colorectal Cancer: High ACAT2 expression correlates with malignancy. Antibodies detected elevated protein levels in GC (gastric cancer) and CRC (colorectal cancer) tissues, linking ACAT2 to cell proliferation and metastasis .
Mechanistic Insights: Knockdown studies using ACAT2 antibodies revealed its interaction with SETD7 and YAP1/TAZ-TEAD1 pathways in cancer progression .
Mouse and Primate Studies: ACAT2 antibodies demonstrated reduced atherosclerosis in Acat2−/− mice, highlighting its role in cholesteryl ester accumulation in lipoproteins .
Human Liver Biopsies: Correlated ACAT2 activity with HDL-C levels, suggesting its impact on cholesterol efflux .
Application | Antibody | Dilution Range | Source |
---|---|---|---|
Western Blotting | 14755-1-AP | 1:2000–1:12000 | |
Immunohistochemistry | ABIN7265364 | 1:20–1:200 | |
Immunoprecipitation | ACAT2 (E1L8V) | 0.5–4.0 µg/mg protein |
The antibody is provided at a concentration of 1mg/ml in a buffer solution containing PBS at a pH of 7.4, 10% Glycerol, and 0.02% Sodium Azide.
ACAT2 is a subtype of Acetyl-coenzyme A acetyltransferase (ACAT), a membrane-bound enzyme that plays important roles in lipid metabolism. It participates in isoleucine degradation, ketolysis, ketogenesis, and fatty acid oxidation . ACAT2 antibodies are crucial research tools that enable the detection, quantification, and localization of ACAT2 in various experimental contexts, particularly in cancer research where ACAT2 has been identified as a potential biomarker and therapeutic target. Recent studies have shown that ACAT2 is significantly upregulated in gastric cancer tissues compared to normal counterparts, and high expression correlates with poor prognosis .
Several methodological approaches are effective for detecting ACAT2 expression:
Immunohistochemistry (IHC): For tissue samples, IHC can effectively detect ACAT2 with optimal antibody dilution of 1:300 (for antibodies like ab131215, Abcam). The protocol involves:
Sectioning paraffin-embedded tissues at 4 μm thickness
Baking at 65°C for 2 hours
Deparaffinizing in xylene and rehydrating through graded alcohol
Placing in 3% hydrogen peroxide
Heating in 1x sodium citrate antigen retrieval buffer (pH 6.0) using a pressure cooker
Blocking with 5% bovine serum albumin
Incubating with primary antibody overnight at 4°C
Incubating with secondary antibody (e.g., MaxVision-HRP) for 1 hour
Visualizing with diaminobenzidine and counterstaining with hematoxylin
Western Blotting: For cell lines, western blotting can be performed using:
Quantitative Real-Time PCR: For mRNA expression:
ACAT2 immunostaining evaluation requires careful scoring and interpretation:
Localization: ACAT2 staining is typically observed in both the cytoplasm and nucleus of cells .
Scoring Method: A comprehensive scoring system involves:
Selecting five high-power fields randomly in each specimen
Examining cytoplasm staining
Calculating immune score = percentage of positive cells × staining intensity
Percentage scoring: 0 (≤5%), 1 (6−25%), 2 (26−50%), 3 (51−75%), 4 (76−100%)
Intensity scoring: 0 (negative), 1 (weak), 2 (moderate), 3 (strong)
Expression Classification:
This standardized approach facilitates reliable comparison across different studies and samples.
ACAT2 antibodies are instrumental in elucidating the molecular mechanisms of ACAT2-mediated cancer progression:
YAP1/TAZ-TEAD1 Signaling Pathway Analysis:
SETD7 Expression Regulation:
Chromatin immunoprecipitation using ACAT2 antibodies can identify direct binding to the SETD7 promoter
Western blotting for SETD7 after ACAT2 manipulation demonstrates regulatory relationships
Research has shown that ACAT2 depletion significantly reduced SETD7 transcription, and ACAT2's pro-tumoral functions were largely dependent on SETD7
EMT Process Evaluation:
Cell Cycle Regulation:
Several methodological approaches can be employed:
Resolving contradictory data requires systematic approaches:
Standardization of Detection Methods:
Use consistent antibody clones and dilutions across studies
Implement uniform scoring systems for immunohistochemistry
Include validated positive and negative controls
Context-Specific Analysis:
Pathway Intersection Analysis:
Meta-analysis Approach:
Systematically combine data from multiple studies
Account for methodological variations
Analyze subgroup differences based on cancer type, stage, and treatment history
Robust experimental design requires comprehensive controls:
For Western Blotting:
Positive control: Cell lines known to express high levels of ACAT2 (e.g., HGC-27, NCI-N87 for gastric cancer; A2780/DDP for ovarian cancer)
Negative control: Cell lines with minimal ACAT2 expression or ACAT2 knockdown samples
Loading control: GAPDH (recommended dilution 1:50000) or other housekeeping proteins
Molecular weight marker to confirm the expected size of ACAT2 (approximately 41 kDa)
For Immunohistochemistry:
For Gene Expression Studies:
A comprehensive validation protocol includes:
Design Phase:
Select target sequences for shRNA design (multiple sequences recommended)
Include non-targeting control shRNA
Consider inducible knockdown systems for temporal control
Knockdown Implementation:
Transduce lentiviral shRNA constructs into target cell lines
Select stable knockdown cell lines using appropriate antibiotics
Optimize MOI (multiplicity of infection) for efficient transduction
Validation Methods:
mRNA Level: qRT-PCR with ACAT2-specific primers
Protein Level: Western blotting with ACAT2 antibody (1:5000 dilution recommended)
Functional Validation: Proliferation assays (CCK-8), colony formation assays, migration assays (wound healing), invasion assays (transwell), and cell cycle analysis by flow cytometry
Phenotypic Confirmation:
To investigate ACAT2's role in ubiquitination:
Co-immunoprecipitation Assays:
Proteasome Inhibition Studies:
Treat cells with proteasome inhibitors (e.g., MG132)
Compare YAP1 protein levels in ACAT2 knockdown vs. control cells
Western blot analysis to detect accumulated ubiquitinated proteins
Ubiquitination Site Mapping:
Mass spectrometry analysis of immunoprecipitated YAP1
Site-directed mutagenesis of potential ubiquitination sites
Evaluate effect of ACAT2 on wild-type vs. mutant YAP1
E3 Ligase Identification:
Screen candidate E3 ligases using siRNA libraries
Validate with co-immunoprecipitation between YAP1 and candidate E3 ligases
Assess how ACAT2 affects these interactions
Evidence-based interpretation requires:
ACAT2's role in metabolism can be investigated through:
Metabolic Pathway Analysis:
Immunoprecipitate ACAT2 to identify interacting metabolic enzymes
Measure acetyl-CoA levels in ACAT2 knockdown vs. control cells
Assess the impact on lipid profiles using lipidomics approaches
Epigenetic Regulation Mechanisms:
Metabolic Inhibitor Combinations:
Test combinations of ACAT2 inhibitors with other metabolic pathway inhibitors
Evaluate synergistic effects on cancer cell viability and drug resistance
Monitor metabolic changes using NMR or mass spectrometry
Advanced methodological approaches include:
Spatial Transcriptomics and Proteomics:
Combine ACAT2 antibody staining with spatial transcriptomics
Map ACAT2 expression patterns within the tumor microenvironment
Correlate with stromal and immune cell markers
3D Organoid Models:
Establish patient-derived organoids maintaining ACAT2 expression patterns
Immunostain for ACAT2 in different organoid regions
Test drug responses in organoids with varying ACAT2 levels
Single-Cell Analysis:
Apply single-cell Western blotting for ACAT2
Combine with single-cell RNA sequencing to correlate protein and mRNA levels
Identify cell subpopulations with distinct ACAT2 expression patterns
Mouse anti-human antibodies are secondary antibodies generated by immunizing mice with human immunoglobulins . These antibodies are widely used in research and diagnostic applications due to their specificity and versatility .
Mouse anti-human antibodies are produced by injecting mice with human immunoglobulins, which stimulates the mouse immune system to produce antibodies against the human proteins . These antibodies are then harvested from the mouse serum and purified using techniques such as immunoaffinity chromatography .
Mouse anti-human antibodies are used in various applications, including:
One important consideration when using mouse anti-human antibodies is the potential for the human anti-mouse antibody (HAMA) response . This response occurs when the human immune system recognizes the mouse antibodies as foreign and mounts an immune response against them . The HAMA response can range from mild allergic reactions to severe complications such as kidney failure .