TNNT2 is a 36 kDa protein expressed in cardiac muscle, essential for calcium-dependent regulation of sarcomere contraction . Mutations in TNNT2 are linked to familial hypertrophic cardiomyopathy (CMD1D) and dilated cardiomyopathy . Recombinant monoclonal antibodies (rMAbs) targeting TNNT2 enable precise detection and quantification of this biomarker in diagnostic and research contexts.
Recombinant methods leverage in vitro transcriptionally-active PCR (TAP) fragments to rapidly generate functional antibodies from single plasma cells, bypassing traditional hybridoma limitations .
*Prices reflect list prices; bulk discounts may apply.
Rapid Antibody Generation:
Cross-Species Reactivity:
Clinical Utility:
The TNNT2 recombinant monoclonal antibody is generated through a meticulous multi-step in vitro process. Initially, TNNT2 antibody genes are extracted from B cells isolated from immunoreactive rabbits. These genes undergo amplification and are cloned into suitable phage vectors. Subsequently, these vectors are introduced into mammalian cell lines to facilitate the production of functional antibodies. The resulting TNNT2 recombinant monoclonal antibody is purified using affinity chromatography. This antibody can be utilized to detect human TNNT2 protein in various applications, including ELISA, IHC, and FC.
TNNT2 plays a pivotal role in cardiac muscle function. It regulates muscle contraction and relaxation by responding to changes in intracellular calcium levels. Its integration within the troponin complex ensures proper heart function and efficient blood pumping throughout the circulatory system.
TNNT2 (cardiac troponin T) is a critical protein in cardiac muscle tissue that regulates muscle contraction and relaxation by sensing changes in intracellular calcium levels. Its role in the troponin complex ensures proper heart function and efficient blood circulation throughout the body . TNNT2 binds to tropomyosin, interlocking them to form a troponin-tropomyosin complex, which helps position tropomyosin on actin and modulates contraction of striated muscle . The significance of TNNT2 in cardiac research stems from its association with several cardiac pathologies, including familial hypertrophic cardiomyopathy and dilated cardiomyopathy . Mutations in the TNNT2 gene can disrupt normal cardiac function, making it an important target for cardiovascular disease research.
TNNT2 recombinant monoclonal antibody generation involves a multi-step in vitro process:
Isolation of B cells from immunoreactive rabbits containing TNNT2 antibody genes
Amplification of these antibody genes
Cloning of the genes into suitable phage vectors
Introduction of vectors into mammalian cell lines for functional antibody production
Purification of the resulting antibodies through affinity chromatography
This approach differs from traditional hybridoma technology used for mouse monoclonal antibody production, which typically involves immunizing mice with an antigen (such as human cTnT or a myofibrillar preparation from ventricle) and fusing antibody-producing B cells with myeloma cells to create hybridomas .
Optimizing TNNT2 antibody cross-species reactivity requires careful consideration of several factors:
Epitope conservation analysis: Before selecting an antibody, analyze the conservation of TNNT2 sequences across species of interest. The central region (amino acids 182-211) of human TNNT2 shows good conservation across some mammalian species .
Antibody selection based on verified reactivity: Choose antibodies explicitly validated for cross-species reactivity. For example:
Titration optimization: When using an antibody across species, perform careful titration experiments to determine optimal working concentrations for each species, as these may differ substantially.
Validation strategy: Confirm specificity through multiple approaches, including western blot analysis in tissue lysates from different species (e.g., mouse heart tissue lysates for antibodies claiming mouse reactivity) .
Preabsorption controls: For polyclonal antibodies, consider preabsorption with the immunizing peptide as a negative control to confirm specificity when working across species.
Non-specific binding is a common challenge when working with TNNT2 antibodies. Implement these strategies to improve specificity:
Optimize blocking conditions:
Use species-appropriate blocking sera (5-10% normal serum from the same species as the secondary antibody)
Consider alternative blocking agents such as BSA (1-5%) or commercial blocking solutions
Extend blocking time to 1-2 hours at room temperature or overnight at 4°C
Antibody dilution optimization:
Tissue preparation considerations:
For formalin-fixed tissues, optimize antigen retrieval methods (heat-induced vs. enzymatic)
Ensure proper fixation times to preserve epitope integrity
For cardiac tissues specifically, consider specialized fixatives that better preserve sarcomeric proteins
Secondary antibody selection:
Additional control experiments:
Include isotype controls (matching the primary antibody's isotype)
Perform peptide competition assays, especially with polyclonal antibodies
Include tissue samples known to be negative for TNNT2 expression
Differentiating between cardiac and skeletal troponin T isoforms requires careful methodological approaches:
Antibody clone selection: Choose antibody clones specifically validated for cardiac troponin T specificity. The RV-C2 clone is known to immunostain cardiac fibers preferentially, although it may also detect some fetal and neonatal skeletal muscle fibers . The UMAB205 clone has been optimized for cardiac troponin T detection .
Complementary detection approaches: Employ multiple detection methods:
Western blotting can differentiate isoforms based on molecular weight differences (cardiac TNNT2 is approximately 35.4-35.8 kDa)
RT-PCR using isoform-specific primers can distinguish between mRNA transcripts
Dual-label immunofluorescence with antibodies against cardiac-specific and skeletal-specific markers
Developmental stage considerations: Be aware that some antibodies may detect both cardiac and skeletal isoforms in developmental contexts. The RV-C2 antibody immunostains both adult cardiac fibers and some fetal/neonatal skeletal muscle fibers .
Tissue-specific positive controls: Include known positive controls:
Adult heart tissue (positive for cardiac TNNT2)
Adult skeletal muscle (negative for cardiac TNNT2)
Fetal skeletal muscle (may show some cardiac TNNT2 expression)
Data interpretation framework: When analyzing results, consider:
The specific epitope recognized by the antibody
Developmental regulation of troponin isoforms
Potential re-expression of fetal isoforms in pathological conditions
Optimizing TNNT2 antibody dilutions requires systematic testing and validation:
Systematic optimization protocol:
Begin with the manufacturer's recommended dilution range
Prepare a series of 2-fold or 3-fold dilutions (minimum 3-5 different concentrations)
Test all dilutions simultaneously on identical samples
Evaluate signal intensity, background, and signal-to-noise ratio
Select the dilution that provides optimal specific signal with minimal background
Sample-specific considerations:
Different tissue types may require different optimal dilutions
Fixation methods significantly impact epitope accessibility and optimal dilution
Expression levels of TNNT2 vary between developmental stages and pathological conditions
Antibody concentration standardization:
Validation markers:
Document optimal dilutions with representative images
Include positive and negative controls at the same dilution
Verify specificity through peptide competition or genetic knockout controls when available
Implementing comprehensive controls is essential for ensuring reliable TNNT2 antibody results:
Positive controls:
Negative controls:
Specificity controls:
Technical controls:
Loading controls for western blots (e.g., GAPDH, β-actin)
Tissue processing controls to verify consistent fixation and processing
Endogenous peroxidase blocking validation for IHC
Comparative controls:
Multiple antibody clones targeting different TNNT2 epitopes
Correlation with mRNA expression data
Comparison with other cardiac markers (e.g., cardiac troponin I)
TNNT2 antibodies enable sophisticated research applications in cardiac development and disease:
Developmental biology applications:
Disease model characterization:
Methodology considerations for developmental studies:
Implementation in regenerative medicine research:
Verification of cardiomyocyte identity in reprogramming experiments
Quality control for engineered cardiac tissues
Assessment of cardiomyocyte purity in therapeutic cell preparations
Monitoring maturation of induced pluripotent stem cell-derived cardiomyocytes
Technical approaches for disease phenotyping:
High-content imaging for morphological characterization
Multiplexed immunofluorescence with other cardiac markers
Quantitative image analysis of sarcomeric organization
Correlation of TNNT2 distribution with functional measurements
Successful multiplexing of TNNT2 antibodies with other cardiac markers requires careful planning:
Antibody selection criteria for multiplexing:
Host species compatibility (select antibodies raised in different species)
Isotype diversity (if using multiple mouse monoclonals, choose different isotypes)
Validated working concentrations for each antibody
Compatible fixation requirements
Recommended marker combinations:
TNNT2 (cardiac troponin T) + TNNI3 (cardiac troponin I) for troponin complex analysis
TNNT2 + α-actinin for sarcomere structural assessment
TNNT2 + connexin-43 for intercalated disc evaluation
TNNT2 + phosphorylated or mutant TNNT2 for post-translational modification studies
Protocol optimization strategies:
Sequential immunostaining for antibodies requiring different antigen retrieval methods
Careful titration of each antibody in the multiplex panel
Block with sera from all secondary antibody host species
Use highly cross-adsorbed secondary antibodies
Consider directly conjugated primary antibodies to avoid cross-reactivity
Advanced multiplexing techniques:
Tyramide signal amplification for sequential detection on the same section
Spectral unmixing for antibodies with overlapping emission spectra
Multi-epitope ligand cartography for sequential antibody detection
Imaging mass cytometry for highly multiplexed protein detection
Data analysis approaches:
Colocalization analysis of TNNT2 with other sarcomeric proteins
Quantification of relative expression levels across markers
Correlation of marker expression with functional parameters
Machine learning classification of cellular phenotypes based on marker patterns
TNNT2 antibodies can be powerful tools for studying post-translational modifications (PTMs):
Approaches for studying TNNT2 phosphorylation:
Combination of pan-TNNT2 antibodies with phospho-specific antibodies
Note that standard antibodies like RV-C2 are not phosphorylation-specific
Western blotting with phosphatase treatment as controls
2D gel electrophoresis to separate phosphorylated isoforms
Phospho-enrichment followed by mass spectrometry
Investigating other PTMs:
O-GlcNAcylation analysis using PTM-specific antibodies alongside TNNT2 detection
Ubiquitination studies through immunoprecipitation with TNNT2 antibodies followed by ubiquitin detection
Acetylation, methylation, and oxidative modifications using specialized detection methods
Proteolytic cleavage products using antibodies recognizing different TNNT2 domains
Methodological considerations:
Rapid tissue/sample handling to preserve labile PTMs
Inclusion of phosphatase/protease inhibitors in extraction buffers
Use of specialized fixation methods that preserve PTMs for immunohistochemistry
Careful selection of extraction buffers to maintain protein-protein interactions
Disease relevance:
Comparison of PTM patterns between healthy and pathological samples
Correlation of PTM changes with contractile dysfunction
Monitoring PTM changes during disease progression
Assessment of therapeutic interventions targeting specific PTMs
Advanced techniques combining TNNT2 antibodies:
Proximity ligation assay to detect TNNT2 interactions with modifying enzymes
FRET-based approaches to study conformational changes induced by PTMs
Super-resolution microscopy to localize PTMs within the sarcomere structure
ChIP-Seq using TNNT2 antibodies to investigate chromatin associations in cardiomyocyte nuclei
TNNT2 antibodies are increasingly valuable for single-cell characterization approaches:
Single-cell protein profiling applications:
Spatial transcriptomics integration:
Combining TNNT2 immunostaining with in situ hybridization techniques
Correlation of protein localization with mRNA expression at single-cell resolution
Multiplex immunofluorescence with RNAscope for simultaneous detection
Spatial mapping of cardiomyocyte subtypes in intact cardiac tissue
Methodological considerations for single-cell approaches:
Optimization of tissue dissociation protocols to preserve epitopes
Careful titration of antibodies for flow cytometry applications
Validation of specificity at the single-cell level
Development of fixation and permeabilization protocols compatible with RNA preservation
Data analysis frameworks:
Dimensionality reduction techniques to visualize heterogeneity
Clustering algorithms to identify cardiomyocyte subpopulations
Trajectory analysis to map developmental or disease progressions
Integration of protein expression data with transcriptomic profiles
Future applications in precision medicine:
Patient-specific cardiomyocyte characterization
Biomarker development for early disease detection
Therapeutic response monitoring at the single-cell level
Identification of resistant cell populations in heart failure
Adapting TNNT2 antibodies for high-throughput screening requires specialized approaches:
Assay development considerations:
Miniaturization of immunoassays for microplate formats
Automation compatibility of staining protocols
Reproducibility assessment across batch preparations
Standardization of positive and negative controls
Z-factor determination to assess assay quality
Screening platform options:
High-content imaging using TNNT2 antibodies for phenotypic screening
AlphaLISA or homogeneous time-resolved fluorescence for solution-based detection
Automated western blotting systems for higher throughput protein analysis
Microfluidic chambers for contractility assessment with TNNT2 immunostaining
Optimization for specific screening applications:
Cardiomyocyte differentiation efficiency screening
Cardiotoxicity assessment of pharmaceutical compounds
Genetic modifier screens using TNNT2 as a phenotypic readout
Drug screens targeting TNNT2 mutations in cardiomyopathy models
Data analysis pipelines:
Machine learning classification of cellular phenotypes
Dose-response relationship modeling
Multiparametric phenotypic profiling
Pattern recognition algorithms for identifying compound mechanisms
Quality control metrics:
Antibody lot-to-lot variation assessment
Positive control trending over screening campaign duration
Intra-plate and inter-plate variability monitoring
Edge effect evaluation and correction strategies
Systematic comparison of TNNT2 antibodies reveals important methodological considerations:
Clone-specific detection characteristics:
Epitope location impacts detection sensitivity in different applications
Central region antibodies (amino acids 182-211) show good cross-species reactivity
N-terminal or C-terminal targeted antibodies may detect different splice variants
Clone UMAB205 was generated using full-length human recombinant TNNT2
Clone RV-C2 was developed using myofibrillar preparation from ventricle
Cross-methodology comparison data:
Western blotting typically shows single bands at 35.4-35.8 kDa for cardiac TNNT2
Immunohistochemistry reveals sarcomeric banding patterns in cardiac tissue
Flow cytometry enables quantitative assessment of expression levels
ChIP applications require antibodies validated for nuclear protein interactions
Performance factors across applications:
Fixation sensitivity varies between clones
Recombinant vs. hybridoma-derived antibodies may show different batch consistency
Monoclonal antibodies provide higher reproducibility but may be more sensitive to epitope modifications
Polyclonal antibodies offer broader epitope recognition but higher background in some applications
Standardization challenges:
Recommendations for comparative studies:
Test multiple antibody clones when establishing new assays
Validate findings with complementary detection methods
Report detailed methodological parameters in publications
Consider both sensitivity and specificity metrics when comparing antibody performance
A comprehensive validation workflow ensures reliable results with TNNT2 antibodies:
Literature assessment:
Review published studies using the specific antibody clone
Evaluate reported applications and limitations
Note any conflicting results or methodological variations
Identify optimal protocols from successful implementations
Initial validation experiments:
Application-specific validation:
Specificity confirmation:
Peptide competition assays
siRNA knockdown or CRISPR knockout controls when available
Comparison with alternative antibody clones targeting different epitopes
Correlation with mRNA expression data
Documentation recommendations:
Record complete antibody information (manufacturer, clone, lot, concentration)
Document all validation experiments with representative images
Maintain detailed protocols for successful applications
Create standardized positive controls for ongoing quality assurance
By following these comprehensive guidelines, researchers can effectively utilize TNNT2 antibodies in their cardiac research programs while ensuring methodological rigor and reproducibility.