INMT antibodies are designed to target the INMT enzyme, which catalyzes the methylation of indolethylamines and related compounds. Key structural and functional insights include:
Antibody Types: Available INMT antibodies include polyclonal (e.g., Novus Biologicals’ rabbit-derived antibody ) and recombinant monoclonal variants (e.g., Cusabio’s product ).
Epitope Specificity: The Novus Biologicals antibody targets a proprietary sequence within the human INMT protein’s central region , while the Cusabio monoclonal antibody is synthesized using phage vectors and mammalian cell lines .
Applications: Validated for Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF) .
INMT is implicated in prostate cancer progression, with studies revealing:
Downregulation in Tumors: INMT expression is reduced in prostate cancer tissues, correlating with lymph node metastasis, Gleason score, and survival rates .
Therapeutic Target: Knockdown of INMT suppresses prostate cancer cell proliferation and promotes apoptosis via MAPK, TGFβ, and Wnt signaling pathways .
Species-Specific Activity: Rat INMT lacks tryptamine-methylating activity, unlike human and rabbit isoforms .
Detoxification Role: INMT detoxifies selenium compounds and regulates tryptamine metabolism, impacting neurochemical pathways .
DMT (N,N-Dimethyltryptamine): An INMT inhibitor shown to suppress castration-resistant prostate cancer (CRPC) growth in vitro and in vivo .
Combination Therapies: INMT knockdown enhances the efficacy of selenium-based compounds (e.g., methaneseleninic acid) by reducing detoxification .
Bi-Isotype Antibodies: IgG–IgA chimeras enhance neutrophil-mediated reactive oxygen species (ROS) production, a strategy adaptable for INMT-targeted therapies .
INMT functions as a thioether S-methyltransferase, exhibiting activity with a variety of thioethers and their corresponding selenium and tellurium compounds. This includes substances like 3-methylthiopropionaldehyde, dimethyl selenide, dimethyl telluride, 2-methylthioethylamine, 2-methylthioethanol, methyl-n-propyl sulfide, and diethyl sulfide. INMT plays a significant role in detoxifying selenium compounds. Additionally, it catalyzes the N-methylation of tryptamine and structurally related compounds.
INMT (Indolethylamine N-methyltransferase) is a transmethylation enzyme that utilizes S-adenosyl-L-methionine to transfer methyl groups to amino groups of small molecule acceptor compounds. It's primarily known for its role in the biosynthesis of N,N-Dimethyltryptamine (DMT), a psychedelic compound found in mammalian brain and other tissues. The protein is approximately 28.9 kilodaltons in mass and may also be known as TEMT or aromatic alkylamine N-methyltransferase.
The importance of INMT in research stems from its involvement in tryptamine metabolism and potential role in endogenous DMT production, which has implications for neuropsychiatric research and understanding of transmethylation pathways in mammals.
INMT antibodies are commonly used in several research applications:
Western Blotting (WB): For detecting INMT protein expression in tissue or cell lysates
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of INMT in samples
Immunohistochemistry (IHC): For visualizing INMT in tissue sections
Immunofluorescence (IF): For cellular localization studies
Immunocytochemistry (ICC): For detection in cultured cells
Flow Cytometry (FCM): For analyzing INMT in cell populations
The choice of application depends on the specific research question being addressed. Most commercial INMT antibodies are validated for at least one or more of these applications.
Selection of an appropriate INMT antibody should consider:
Species reactivity: Ensure the antibody recognizes INMT from your species of interest. There is significant sequence divergence between species - rabbit and human INMT are 89% homologous, while rat INMT is only 57% homologous with either.
Application validation: Verify the antibody has been validated for your specific application (WB, ELISA, IHC, IF, etc.)
Clonality:
Polyclonal antibodies offer broader epitope recognition but less specificity
Monoclonal antibodies provide higher specificity but may be less sensitive
Epitope region: Some antibodies target specific regions (N-terminal, C-terminal, or internal domains) which may affect detection depending on protein modifications or interactions
Format: Consider whether you need conjugated (HRP, FITC, biotin) or unconjugated antibodies based on your detection system
Supporting data: Review any validation data, including Western blots or immunofluorescence images provided by the manufacturer.
For optimal Western blotting with INMT antibodies:
Sample preparation:
Use fresh tissue/cell lysates with appropriate protease inhibitors
INMT is approximately 28.9 kDa; adjust gel percentage accordingly (10-12% typically works well)
Antibody dilution:
Blocking and incubation conditions:
Block with 5% non-fat dry milk or BSA in TBST
Incubate primary antibody overnight at 4°C
Use compatible secondary antibody (anti-rabbit for rabbit-derived antibodies)
Controls:
Positive control: Human adrenal gland tissue or cell lines known to express INMT
Negative control: Tissue from INMT knockout models or cells with confirmed absence of INMT
Detection:
For successful immunofluorescence using INMT antibodies:
Sample preparation:
For fixed cells: 4% paraformaldehyde for 15-20 minutes
For tissue sections: Consider both frozen and paraffin-embedded sections depending on epitope sensitivity
Antibody dilution and incubation:
Background reduction:
Thorough blocking with 5-10% normal serum from the same species as the secondary antibody
Include 0.1-0.3% Triton X-100 for permeabilization
Consider autofluorescence quenching methods if using tissues with high autofluorescence
Subcellular localization:
INMT is primarily cytoplasmic, as confirmed in multiple cell lines
Co-staining with organelle markers may help determine precise subcellular localization
Controls and counterstaining:
Non-specific binding is a common issue with antibodies. For INMT antibodies:
Verify antibody specificity:
Optimize blocking conditions:
Try different blocking agents (BSA, normal serum, commercial blockers)
Increase blocking time to 1-2 hours at room temperature
Adjust antibody concentration:
Titrate antibody to determine optimal concentration
Too high concentration can lead to non-specific binding
Modify washing protocol:
Increase washing duration and number of washes
Add 0.1-0.5% Tween-20 to wash buffer to reduce non-specific interactions
Pre-absorb the antibody:
If non-specific binding persists, consider pre-absorbing with tissues or cell lysates from species without target protein
Validating antibody specificity is crucial for reliable results:
Knockout/knockdown controls:
Peptide competition assay:
Pre-incubate antibody with excess immunizing peptide
Specific signals should be blocked by the competing peptide
Multiple antibody validation:
Use multiple antibodies targeting different epitopes of INMT
Consistent results across antibodies increase confidence in specificity
Recombinant protein controls:
Use purified recombinant INMT as positive control
Test against recombinant related proteins to check cross-reactivity
Mass spectrometry validation:
Use immunoprecipitation followed by mass spectrometry to confirm antibody is pulling down INMT
Particularly useful for validating antibodies for immunoprecipitation applications
Research has revealed significant species differences in INMT activity that can be explored using antibodies:
Comparative expression analysis:
Use species cross-reactive antibodies to compare INMT expression across species
Western blotting and immunohistochemistry can reveal distribution differences
Functional correlation studies:
Sequence-function relationships:
Tissue distribution mapping:
Map INMT tissue distribution across species using immunohistochemistry
Compare with functional data to identify correlations between expression and activity
Alternative pathway investigation:
To investigate INMT's role in DMT biosynthesis:
Co-localization studies:
Use dual immunofluorescence to co-localize INMT with tryptamine synthesis enzymes
Examine subcellular localization to identify compartmentalization of the biosynthetic pathway
Activity correlation:
Correlate INMT protein levels (by immunoblotting) with DMT production (by mass spectrometry)
Compare across tissues and experimental conditions
Pathway mapping using genetic models:
Biochemical isolation:
Use INMT antibodies for immunoprecipitation to isolate active enzyme complexes
Analyze associated proteins to identify partners in the DMT biosynthetic pathway
Single-cell analysis:
Apply immunofluorescence in conjunction with RNA-FISH to correlate INMT protein with mRNA at single-cell resolution
Identify specific cell populations involved in DMT biosynthesis
For comprehensive analysis of INMT in complex biological systems:
Multi-color flow cytometry:
Combine INMT antibodies with markers for cell types, activation states, and other pathway components
Useful for identifying specific cell populations expressing INMT
Mass cytometry (CyTOF):
Use metal-conjugated INMT antibodies for high-dimensional analysis
Can be combined with dozens of other parameters for comprehensive phenotyping
Multiplex immunohistochemistry:
Employ spectral unmixing or sequential staining approaches
Visualize INMT alongside tissue architecture and other protein markers
Spatial transcriptomics integration:
Combine INMT immunostaining with spatial transcriptomics
Correlate protein expression with local gene expression patterns
Live-cell imaging applications:
Use fluorescently-labeled Fab fragments of INMT antibodies
Track dynamic changes in INMT localization in response to stimuli
When developing INMT as a potential biomarker:
Standardization of detection methods:
Establish consistent protocols for quantitative measurement
Validate across different antibody lots and detection platforms
Tissue-specific considerations:
INMT expression varies across tissues, requiring tissue-specific validation
Consider using tissue microarrays for systematic evaluation
Sensitivity and specificity optimization:
Determine limits of detection and quantification
Establish normal reference ranges in relevant populations
Correlation with clinical parameters:
Sample collection and processing:
Standardize pre-analytical variables (collection, storage, processing)
Validate stability of INMT under different storage conditions
For immunotoxicology investigations involving INMT:
Effects on enzyme activity:
Use antibodies to determine if toxicant exposure alters INMT expression
Correlate with functional changes in methylation capacity
Tissue distribution alterations:
Assess changes in INMT distribution following toxicant exposure
Map co-localization with indicators of cellular stress or damage
Mechanisms of toxicity:
Use proximity ligation assays with INMT antibodies to detect altered protein interactions
Investigate whether toxicants affect INMT's association with cellular structures
Adaptation responses:
Screening applications:
Develop high-content screening approaches using INMT antibodies
Incorporate into multiplexed assays for toxicant screening
| Detection Method | Sensitivity | Specificity | Sample Requirements | Key Advantages | Limitations |
|---|---|---|---|---|---|
| Western Blot | Moderate | High | Protein lysates (10-50 μg) | Size verification, semi-quantitative | Time-consuming, not suitable for tissue localization |
| ELISA | High | Moderate-High | Purified samples, serum (1-100 μl) | Quantitative, high-throughput | No size verification, potential cross-reactivity |
| Immunohistochemistry | Moderate | Moderate | Fixed tissue sections | Spatial distribution, cellular context | Semi-quantitative, fixation artifacts |
| Immunofluorescence | Moderate-High | Moderate-High | Fixed cells/tissues | Subcellular localization, co-localization studies | Photobleaching, autofluorescence |
| Flow Cytometry | High | Moderate-High | Single-cell suspensions | Single-cell resolution, multi-parameter | Limited to cells in suspension |
| Mass Spectrometry | Very High | Very High | Purified protein/peptides | Absolute identification, post-translational modifications | Expensive, technically demanding |
This comparative analysis can help researchers select the most appropriate method based on their specific research questions and available resources .
For successful immunoprecipitation of INMT:
Antibody selection:
Choose antibodies specifically validated for immunoprecipitation
Consider using monoclonal antibodies for higher specificity
Lysis conditions:
Use mild detergents (e.g., 1% NP-40 or 0.5% CHAPS) to preserve protein interactions
Include protease and phosphatase inhibitors to prevent degradation
Pre-clearing strategy:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Consider using isotype control antibodies for comparison
Antibody immobilization:
Either pre-bind antibodies to beads or add directly to lysate
For co-immunoprecipitation studies, consider crosslinking antibody to beads
Validation approaches:
Confirm specificity using INMT knockout samples as negative controls
Perform reverse immunoprecipitation with interaction partners
Apply mass spectrometry for unbiased verification of pull-down specificity
This methodological approach is similar to techniques used for other immunological research applications .