TNNI3 (1-210) is a non-glycosylated polypeptide produced recombinantly in E. coli. Key characteristics include:
Sequence: 210 amino acids (AA) with a His-tag fusion at the N-terminus for purification .
Molecular Weight: 26.4 kDa (recombinant variant) ; native form: ~24 kDa .
Physical Formulation: Supplied in 20 mM Tris-HCl (pH 7.5), 0.1 M NaCl, 30% glycerol, 0.1 mM PMSF, and 1 mM DTT .
Amino Acid Sequence Highlights:
The N-terminal extension (AA 1–30) contains a PKA phosphorylation motif (RRRSS), critical for calcium sensitivity regulation . The C-terminal region (AA 150–210) binds tropomyosin and stabilizes the troponin-tropomyosin complex in low-calcium states .
TNNI3 regulates contraction via calcium-dependent interactions with troponin T (TNNT2) and C (TNNC1). Key mechanisms include:
Calcium Sensitivity Modulation: Binds TNNC1 to inhibit actomyosin ATPase activity in diastole .
Post-Translational Modifications:
Binding Activity: Recombinant TNNI3 binds TNNT2 in a dose-dependent manner, as demonstrated by ELISA assays .
TNNI3 mutations are implicated in inherited cardiomyopathies.
Arg192Cys (R192C): A de novo mutation causing restrictive cardiomyopathy with preserved ejection fraction .
D190Y: Linked to restrictive and hypertrophic cardiomyopathy in Indian populations .
TNNI3 is a critical tool in cardiovascular research:
Structural Studies: AlphaFold predictions reveal interaction domains with TNNT2 and tropomyosin .
Chimeric Constructs: Engineered TNNI3 variants are tested for therapeutic efficacy in cardiac disease models .
Biomarker Utility: Serum TNNI3 levels detect myocardial injury with high specificity .
Parameter | TNNI3 (Cardiac) | TNNI1 (Slow Skeletal) | TNNI2 (Fast Skeletal) |
---|---|---|---|
Gene Location | 19q13.4 | 1q31.3 | 11p15.5 |
Exons | 8 | 9 | 8 |
Amino Acids | 210 | 187 | 182 |
Molecular Weight (kDa) | 24.0 | 21.7 | 21.3 |
Tissue Specificity | Adult heart | Slow-twitch muscles | Fast-twitch muscles |
Isoelectric Point (pI) | 9.87 | 9.59 | 8.74 |
Troponin I (TnI) is a protein integral to the troponin complex, which plays a crucial role in the regulation of striated muscle contraction. Within this complex, TnI functions as the inhibitory subunit, preventing the interaction between actin and myosin, thus facilitating muscle relaxation. The TNNI3 gene specifically encodes the cardiac isoform of TnI, and its expression is restricted to cardiac muscle tissues. Genetic alterations in the TNNI3 gene have been linked to familial hypertrophic cardiomyopathy type 7 (CMH7) and familial restrictive cardiomyopathy (RCM), underscoring its importance in maintaining normal heart function.
Recombinant Human TNNI3, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein spans 233 amino acids, encompassing residues 1-210 of the TNNI3 sequence, with an additional 23 amino acid His-tag fused at the N-terminus. It possesses a molecular weight of 26.4 kDa and is purified to homogeneity using proprietary chromatographic techniques.
The TNNI3 protein solution is provided at a concentration of 0.25mg/ml. The storage buffer consists of 30% glycerol, 20mM Tris-HCl (pH 7.5), 0.1M NaCl, 0.1mM PMSF, and 1mM DTT.
The purity of the TNNI3 protein is greater than 90.0%, as determined by SDS-PAGE analysis.
MGSSHHHHHH SSGLVPRGSH MGSMADGSSD AAREPRPAPA PIRRRSSNYR AYATEPHAKK KSKISASRKL QLKTLLLQIA KQELEREAEERRGEKGRALS TRCQPLELAG LGFAELQDLC RQLHARVDKV DEERYDIEAK VTKNITEIAD LTQKIFDLRG KFKRPTLRRV RISADAMMQALLGARAKESL DLRAHLKQVK KEDTEKENRE VGDWRKNIDA LSGMEGRKKK FES
Cardiac Troponin I (TNNI3) is an integral inhibitory protein in cardiac muscle that forms part of a complex with troponin C (TnC) and troponin T (TnT). The protein functions as a key regulator of the contraction-relaxation function of cardiac muscle and is expressed strongly and specifically in cardiac muscle tissues .
The amino acid sequence 1-210 represents nearly the complete protein structure of human cardiac troponin I. This sequence contains critical functional domains including:
The inhibitory region that interacts with actin
Calcium-responsive regulatory elements
Binding sites for troponin C and troponin T interactions
Regions subject to post-translational modifications that regulate contractile function
The protein's structure enables its role in the troponin-tropomyosin complex, where it regulates muscle contraction through calcium-dependent interactions. When calcium binds to specific sites on troponin C, it induces structural changes that release the inhibitory region of TNNI3 from actin, thereby enabling myosin attachment and muscle contraction .
Several expression systems have been employed for producing recombinant TNNI3, each with specific advantages for different research applications:
Expression System | Advantages | Typical Purity | Applications |
---|---|---|---|
Escherichia coli (E. coli) | High yield, cost-effective, simpler purification | >90-97% | Western blotting, SDS-PAGE, immunogen preparation, positive controls |
Wheat germ | Enhanced folding for complex proteins | Not specified | Western blotting, ELISA, AP, AA assays |
Escherichia coli is the most commonly used expression system for producing recombinant human TNNI3 (1-210) with His tags for easy purification. This system provides high yields of protein that can achieve >90% purity as measured by SDS-PAGE gel analysis .
For experimental applications requiring native-like protein folding, wheat germ expression systems may be preferable, though with potentially lower yields. Production in E. coli typically employs codon optimization, IPTG induction, and purification via nickel affinity chromatography to isolate the His-tagged protein .
Detection of TNNI3 in research and clinical settings employs several methodological approaches:
ELISA-Based Detection:
The Human TNNI3 solid-phase sandwich ELISA is designed to measure target protein bound between matched antibody pairs. This method involves:
Pre-coating wells with target-specific capture antibody
Adding samples, standards, or controls to bind to the immobilized antibody
Forming a sandwich with the addition of a detector antibody
Adding substrate solution that produces measurable signal proportional to TNNI3 concentration
Western Blotting (WB):
Western blotting remains a standard technique for validating TNNI3 expression, with recombinant TNNI3 proteins serving as positive controls. This approach allows for size verification and semi-quantitative analysis .
SDS-PAGE Analysis:
SDS-PAGE is commonly used for purity assessment of recombinant TNNI3, with most commercial preparations achieving >90% purity as visualized by Coomassie blue staining .
Each method offers different advantages for sensitivity, specificity, and throughput, making method selection crucial for experimental design.
TNNI3 mutations have been significantly implicated in cardiac pathologies, particularly hypertrophic cardiomyopathy (HCM) and atrial fibrillation (AF). Whole-exome sequencing and targeted screening approaches have identified several pathogenic variants:
Key Pathogenic Variants:
Mutation | Associated Condition | Population | Penetrance | Reference |
---|---|---|---|---|
R186Q | HCM and AF | Chinese, French, Caucasian, Asian | Incomplete, age-dependent | Richard et al. 2003; Mogensen et al. 2004 |
E64G | AF | Chinese | Unknown | Wang et al. 2015 |
M154L | AF | Chinese | Unknown | Wang et al. 2015 |
E187G | AF | Chinese | Unknown | Wang et al. 2015 |
D196G | AF | Chinese | Unknown | Wang et al. 2015 |
The R186Q mutation demonstrates incomplete, age-dependent penetrance. In one Chinese family study, 9 members carried the R186Q mutation, but 3 were unaffected by either HCM or AF. This variable expressivity suggests complex genotype-phenotype relationships and potential genetic or environmental modifiers .
Mechanistically, these mutations likely alter the regulatory function of troponin I in cardiac contraction, affecting calcium sensitivity, energy utilization, or interaction with other sarcomeric proteins. The identification of TNNI3 variants in AF patients without structural heart disease (lone AF) suggests that altered contractile function may directly affect atrial electrophysiology .
Comprehensive identification and validation of TNNI3 variants requires a multi-method approach:
Sequencing Technologies:
Whole-exome sequencing (WES) provides comprehensive coverage for identifying novel variants across the entire coding region
High-resolution melting curve analysis offers a rapid screening method for known hotspot regions
Direct DNA sequence analysis remains the gold standard for confirmation
Validation Workflow:
Initial Discovery: Use WES in family cohorts or selected patient populations
Confirmation: Perform Sanger sequencing of candidate variants
Population Screening: Screen larger cohorts of patients and controls using targeted approaches
Co-segregation Analysis: Test whether the variant co-segregates with disease in families
In Silico Analysis: Assess conservation, predicted functional impact, and absence from population databases
Functional Studies: Express mutant proteins for biochemical and cellular assays
When identifying rare variants like those in TNNI3, it's critical to note that genome-wide association studies (GWAS) often miss these associations due to their extremely low minor allele frequencies. This limitation highlights the advantage of next-generation sequencing approaches for identifying rare pathogenic variants .
Researchers face significant challenges when analyzing TNNI3 variant data across different genomic databases due to issues with completeness and concordance:
Completeness Challenges:
Genomic databases vary considerably in their coverage of TNNI3 variants. Completeness can be evaluated at two levels:
Schema Level: Whether the database provides all relevant fields (variant details, clinical significance, population frequency)
Data Level: Whether the database contains all known variants for a specific disorder
For comprehensive TNNI3 research, multiple databases should be consulted, as no single source contains all variants.
Concordance Issues:
Discordant interpretations of the same TNNI3 variant can occur between databases, which threatens result correctness. Concordance can be assessed through:
Interpretation Approach: Comparing whether multiple sources agree on pathogenicity classifications
Statistical Association Approach: Comparing statistical significance metrics (p-values, confidence intervals)
Reconciliation Methodology:
To address these challenges, researchers should:
Query multiple databases including ClinVar, Ensembl, GWAS Catalog, LOVD, and cardiology-specific databases
Calculate completeness metrics to understand database coverage limitations
Assess concordance for variants of interest across all available sources
Prioritize concordant findings but carefully evaluate discordant ones through additional functional studies
Consider using metrics like Completeness at the Data Level (CDL) to quantify information coverage
The Human Cardiac Troponin I (TNNI3) ELISA requires careful optimization for accurate quantification in cardiac research:
Sample Preparation Considerations:
Human serum and plasma require standardized collection protocols to prevent proteolytic degradation
Cell culture medium samples may need concentration depending on expression levels
Proper sample dilution series is essential for accurate quantification within the assay's linear range
Assay Optimization Parameters:
Antibody concentrations and incubation times should be validated for each experimental setup
Temperature control during incubation steps significantly impacts assay performance
Washing protocols must be optimized to reduce background without losing sensitivity
Quality Control Requirements:
Each manufactured ELISA kit batch should undergo validation for sensitivity, specificity, precision, and lot-to-lot consistency
Standard curves must be prepared fresh for each assay run
Both positive and negative controls should be included in every experiment
Data Analysis Approach:
Four-parameter logistic curve fitting is recommended for standard curve analysis
The lower limit of detection should be experimentally determined for each laboratory's conditions
Spike and recovery experiments help validate matrix effects in complex biological samples
These methodological considerations are critical for obtaining reliable TNNI3 measurements, particularly when comparing clinical samples or evaluating experimental interventions.
Investigating the functional consequences of TNNI3 mutations requires carefully designed experimental approaches:
In Vitro Contractile Protein Studies:
Express and purify wild-type and mutant TNNI3 (1-210) proteins using bacterial expression systems
Reconstitute troponin complex with recombinant TnC and TnT
Perform calcium sensitivity assays using skinned muscle fibers or reconstituted filaments
Measure force development and ATPase activity under varying calcium concentrations
Cellular Models:
Generate cardiomyocyte models expressing wild-type or mutant TNNI3 using:
Transfected cell lines (limited physiological relevance)
Primary cardiomyocytes (challenging but physiologically relevant)
iPSC-derived cardiomyocytes (patient-specific or engineered mutations)
Assess contractile parameters, calcium handling, and sarcomere organization
Evaluate protein-protein interactions using co-immunoprecipitation or proximity ligation assays
Animal Models:
Generate transgenic or knock-in mice expressing human TNNI3 mutations
Perform comprehensive cardiac phenotyping including:
Echocardiography for structural and functional assessment
Electrocardiography for arrhythmia detection
Histological analysis for tissue remodeling
Isolated cardiomyocyte studies for contractile properties
Evaluate age-dependent penetrance of phenotypes to model human disease progression
Translational Validation:
Correlate experimental findings with clinical data from mutation carriers, including:
Age of onset
Phenotype severity
Response to treatments
Long-term outcomes
This multi-level experimental approach provides mechanistic insights into how TNNI3 mutations disrupt normal cardiac function and lead to pathologies like atrial fibrillation and hypertrophic cardiomyopathy .
Current TNNI3 research faces several important limitations that present opportunities for future investigation:
Data Integration Challenges:
The fragmentation of TNNI3 variant data across multiple databases creates significant challenges for comprehensive analysis. Future research should focus on developing integrated databases and analytical tools specifically for cardiac-related genes that incorporate completeness and concordance metrics .
Phenotypic Heterogeneity:
The same TNNI3 mutation (e.g., R186Q) can produce varying clinical phenotypes with incomplete penetrance. Further research is needed to identify genetic modifiers and environmental factors that influence disease expression .
Functional Characterization Gap:
While multiple TNNI3 variants have been identified in cardiac patients, functional characterization remains limited for many variants. High-throughput functional screening methods are needed to classify variants of uncertain significance.
Therapeutic Translation:
Despite advances in understanding TNNI3's role in cardiac disease, targeted therapeutic approaches remain underdeveloped. Future research should focus on:
Developing small molecules that can modulate mutant TNNI3 function
Exploring gene editing approaches to correct pathogenic mutations
Investigating RNA-based therapeutics to modulate TNNI3 expression levels
Methodological Standardization: Different laboratories employ varying methods for TNNI3 detection and functional analysis, complicating cross-study comparisons. Standardized protocols for TNNI3 expression, purification, and functional characterization would significantly advance the field.
Cardiac Troponin-I is a critical protein in the regulation of cardiac muscle contraction. It is part of the troponin complex, which includes Troponin T and Troponin C, and plays a pivotal role in the calcium-mediated regulation of muscle contraction. The specific form, Human Recombinant Cardiac Troponin-I (1-210 a.a.), is a truncated version of the full-length protein, expressed in Escherichia coli for research and diagnostic purposes.
Troponin-I binds to actin in thin myofilaments to hold the actin-tropomyosin complex in place, preventing myosin from binding to actin in relaxed muscle . This inhibition is crucial for muscle relaxation and contraction cycles. The recombinant form, spanning amino acids 1 to 210, retains the essential functional domains necessary for its inhibitory role.
The recombinant Human Cardiac Troponin-I (1-210 a.a.) is typically expressed in Escherichia coli. This bacterial expression system is favored due to its simplicity, cost-effectiveness, and ability to produce large quantities of protein. The protein is then purified to a high degree of purity (>90%) using techniques such as SDS-PAGE .
Cardiac Troponin-I is a highly specific biomarker for myocardial injury. Its levels in the blood rise significantly within a few hours of myocardial infarction, making it an essential tool in the diagnosis of acute coronary syndromes . The recombinant form is used in various assays to detect and quantify Troponin-I levels in clinical samples.
Recombinant Human Cardiac Troponin-I (1-210 a.a.) is widely used in research to study the molecular mechanisms of cardiac muscle contraction and regulation. It is also employed in the development and validation of diagnostic assays for cardiac biomarkers.