TNNI3 Human Native

Cardiac Troponin-I Human
Shipped with Ice Packs
In Stock

Description

Clinical Significance in Cardiovascular Diseases

Mutations in TNNI3 are linked to:

  • Familial hypertrophic cardiomyopathy (CMH7)

  • Restrictive cardiomyopathy (RCM)

Case Study Highlight: A de novo TNNI3 mutation (c.574C>T, p.Arg192Cys) caused severe RCM in a 2-year-old patient, necessitating heart transplantation . This mutation disrupts cTnI’s interaction with tropomyosin, leading to diastolic dysfunction .

Diagnostic Biomarker

  • Elevated cTnI levels in serum indicate myocardial injury, with >95% specificity for cardiac events .

  • Native TNNI3 is used to study calcium-mediated contraction mechanisms in cardiac tissue .

Pathogenic Mutations

MutationPhenotypePopulationClinical Impact
p.Arg192CysRCMChineseDiastolic dysfunction, requires transplantation .
p.Asp190TyrRCM with mild hypertrophyIndianReduced systolic function .
p.Ser150TrpDiabetic cardiomyopathyMouse modelsImpaired contractility due to O-GlcNAcylation .

Therapeutic Research

  • Native TNNI3-expressing cardiomyocytes model diseases like myocardial stunning .

  • Targeting TNNI3 phosphorylation sites may improve outcomes in heart failure .

Evolutionary Conservation

The N-terminal extension of cTnI is ancestral to vertebrates but lost in teleost fish, highlighting its role in advanced cardiac regulation . Structural conservation across mammals supports its critical function .

Product Specs

Description
TNNI3 Native produced in Human heart tissue is a full length protein with additional amino acid residues on its N terminus not present in the skeletal form, making it a promising analyte for indicating cardiac specificity. TNNI3 Native is purified by proprietary chromatographic technique.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
TNNI3 was lyophilized from 0.01M HCl.
Stability
Lyophilized Cardiac Troponin-I is stable at room temperature for 3 weeks but should be stored desiccated below -18°C. Upon reconstitution, TNNI3 should be stored at 4°C for 2-7 days and below -18°C for future use. For long term storage, add a carrier protein (0.1% HSA or BSA). Avoid freeze-thaw cycles.
Solubility
Reconstitute the lyophilized TNNI3 in Tris/urea buffer (20mM Tris, pH 7.5, 7M urea, 5mM EDTA, 15mM 2-mercaptoethanol) at a concentration not less than 100µg/ml. This solution can be further diluted with other aqueous solutions.
Purity
Greater than 98.0% as determined by SDS-PAGE.
Synonyms

Troponin I cardiac muscle, Cardiac troponin I, TNNI3, TNNC1, CMH7, RCM1, cTnI, CMD2A, MGC116817.

Source

Human heart tissue.

Q&A

What is TNNI3 and what is its role in cardiac function?

TNNI3 (Troponin I Type 3) is one of three isoforms of Troponin I that is exclusively expressed in the heart. It functions as a critical regulatory protein in the cardiac contractile apparatus, inhibiting actomyosin ATPase activity in the absence of calcium. When calcium binds to the troponin complex, TNNI3 undergoes conformational changes that permit actin-myosin interaction, enabling contraction. TNNI3 is particularly crucial for proper relaxation and diastolic function, with mutations in this gene frequently associated with restrictive cardiomyopathy (RCM) .

The protein's unique structural features allow it to respond precisely to calcium fluctuations, making it essential for the beat-to-beat regulation of cardiac contraction and relaxation cycles. Experimental models have consistently demonstrated that alterations in TNNI3 function directly impact cardiac relaxation properties, highlighting its fundamental importance in heart physiology.

How does TNNI3 expression change during cardiac development?

During cardiac development, a critical isoform switch occurs wherein TNNI3 is upregulated during the transition from fetal to neonatal and postnatal periods, eventually replacing TNNI1 (the fetal isoform) as the sole troponin I isoform in the adult heart . This developmental transition represents a key maturation marker in cardiomyocytes and has significant functional implications.

The TNNI1-to-TNNI3 switch correlates with changes in calcium sensitivity, contractile properties, and response to regulatory mechanisms such as adrenergic stimulation. This transition is particularly relevant when studying induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), as these cells typically exhibit an immature phenotype similar to fetal cardiomyocytes but undergo maturation processes associated with increased TNNI3 expression .

What experimental techniques are most reliable for quantifying TNNI3 expression?

Reliable quantification of TNNI3 requires a combination of complementary techniques:

  • Protein-level analysis:

    • Western blotting with isoform-specific antibodies

    • Mass spectrometry for absolute quantification

    • Immunofluorescence for spatial distribution assessment

  • Transcript analysis:

    • RT-qPCR with carefully validated primers

    • RNAseq for comprehensive transcriptomic profiling

    • Droplet digital PCR for absolute quantification

  • Functional correlates:

    • Calcium sensitivity assays

    • Phosphorylation state assessment

    • Force-calcium relationship measurements

For developmental studies, researchers should implement time-course experiments comparing TNNI3:TNNI1 ratios at multiple differentiation timepoints. Serial measurements beginning at day 20 post-differentiation and continuing through days 40, 60, and 90 provide the most informative dataset for assessing cardiomyocyte maturation status .

What types of cardiomyopathies are associated with TNNI3 mutations?

TNNI3 mutations have been implicated in multiple forms of hereditary cardiomyopathy, with particularly strong associations with:

Cardiomyopathy TypeApproximate Prevalence of TNNI3 MutationsCommon Mutation LocationsPredominant Functional Effect
Restrictive Cardiomyopathy (RCM)5-10% of genetic casesR170, R192, K178Impaired relaxation
Hypertrophic Cardiomyopathy (HCM)3-5% of genetic casesR145, R162, P82Increased calcium sensitivity
Dilated Cardiomyopathy (DCM)1-2% of genetic casesK36, N185, D196Decreased contractility

The R170W mutation specifically has been documented in cases of early childhood-onset RCM, with impaired relaxation being the hallmark phenotype . This mutation occurs in a highly conserved region of the protein that interacts with other components of the troponin complex, thereby disrupting normal cardiac relaxation mechanisms.

What methodologies are most effective for studying TNNI3 mutations in relation to restrictive cardiomyopathy?

A comprehensive approach to studying TNNI3 mutations in RCM should incorporate:

  • Patient-derived iPSC models:

    • Generate iPSCs from patients with confirmed TNNI3 mutations

    • Create isogenic control lines using CRISPR-Cas9 gene editing

    • Differentiate into high-purity cardiomyocyte populations

  • Advanced tissue engineering:

    • Develop engineered heart tissue (EHT) constructs that recapitulate three-dimensional architecture

    • EHT models have proven particularly advantageous for studying the impaired relaxation phenotype characteristic of RCM

    • Implement biomechanical conditioning to enhance tissue maturation

  • Functional assessments:

    • Contractility measurements (force generation, contraction velocity)

    • Relaxation parameters (relaxation time, passive tension)

    • Calcium transient recordings with high temporal resolution

  • Molecular interventions:

    • Viral-mediated gene delivery of wild-type TNNI3

    • siRNA-based knockdown approaches

    • Small molecule compound screening

Research has demonstrated that engineered heart tissues can precisely recapitulate the impaired relaxation of RCM in vitro, providing a valuable platform for mechanistic studies and therapeutic development .

How do researchers differentiate between pathogenic and benign TNNI3 variants?

Distinguishing pathogenic from benign TNNI3 variants requires a multi-faceted approach:

  • Computational prediction:

    • Evolutionary conservation analysis across species

    • Structural modeling of troponin complex interactions

    • Machine learning algorithms trained on known pathogenic variants

  • Functional characterization:

    • Force-calcium relationship measurements in reconstituted systems

    • Protein stability and binding affinity assessments

    • Phosphorylation dynamics evaluation

  • Clinical correlation:

    • Family segregation analysis

    • Penetrance assessment in carriers

    • Phenotype-genotype correlation studies

  • Population frequency:

    • Examination in population databases (gnomAD, ExAC)

    • Ethnic-specific frequency considerations

    • Allele frequency comparison with disease prevalence

Variants affecting highly conserved residues at protein-protein interaction interfaces, particularly in calcium-sensing regions, warrant careful evaluation regardless of computational predictions.

What considerations should be made when designing iPSC-CM models to study TNNI3 mutations?

When designing iPSC-CM models for TNNI3 mutation studies, researchers should address:

  • Maturation status:

    • Standard differentiation protocols produce cardiomyocytes with fetal-like characteristics, including incomplete TNNI1-to-TNNI3 isoform switching

    • Implement maturation-enhancing strategies (electrical stimulation, mechanical loading, T3 supplementation)

    • Validate maturation using comprehensive markers, including TNNI3:TNNI1 ratio

  • Genetic background:

    • Generate isogenic controls to eliminate confounding genetic variations

    • Consider multiple patient lines to account for genetic modifier effects

    • Validate findings in different genetic backgrounds when possible

  • Experimental endpoints:

    • Include both structural and functional assessments

    • Measure calcium handling parameters (transient amplitude, decay kinetics)

    • Assess contractile and relaxation parameters using appropriate platforms

  • Tissue architecture:

    • 3D tissue constructs better recapitulate physiological conditions than 2D monolayers

    • Engineered heart tissues specifically demonstrate superior capacity for detecting relaxation abnormalities in TNNI3-associated RCM

    • Consider cell alignment and electrical coupling in tissue design

The cardiac maturation obstacle in experimental disease models using iPSC-CMs may account for the historical lack of in vitro studies of diseases caused by TNNI3 abnormalities .

How can engineered heart tissue (EHT) models be optimized for TNNI3 mutation studies?

Optimizing EHT models for TNNI3 mutation research requires:

  • Mechanical considerations:

    • Implement defined substrate stiffness matching native myocardium

    • Apply cyclic mechanical strain during tissue formation

    • Utilize pillar-based systems for continuous force monitoring

  • Cellular composition:

    • Incorporate cardiac fibroblasts at physiological ratios (10-20%)

    • Consider endothelial cell co-culture for paracrine signaling

    • Ensure high cardiomyocyte purity (>90%) with minimal non-cardiac contamination

  • Maturation enhancement:

    • Extend culture duration beyond 4 weeks

    • Apply electrical field stimulation with physiological pacing protocols

    • Include maturation-promoting factors (T3, insulin, fatty acids)

  • Functional assessment:

    • Implement high-speed video recording for contractile analysis

    • Measure force generation under varying preload/afterload conditions

    • Assess frequency-dependent force generation and relaxation

Research has demonstrated that mature EHT constructs represent an advantageous model for studying the RCM phenotype of impaired relaxation associated with TNNI3 mutations .

What are the limitations of current TNNI3 animal models and how can they be addressed?

Current TNNI3 animal models face several limitations:

  • Species-specific differences:

    • Murine models exhibit different calcium handling properties

    • Cardiac contraction rates differ significantly from humans

    • Response to pharmacological interventions shows species variation

  • Developmental compensation:

    • Embryonic lethal phenotypes with complete knockout

    • Compensatory mechanisms may mask acute effects

    • Heterozygous models may incompletely recapitulate human disease

  • Technical challenges:

    • Difficulty in producing tissue-specific conditional knockins

    • Variation in phenotype severity with genetic background

    • Limited throughput for therapeutic screening

Addressing these limitations requires:

  • Developing inducible expression systems for temporal control

  • Implementing human iPSC-derived xenografts in immunodeficient hosts

  • Utilizing large animal models for translational validation

  • Combining multiple model systems to validate findings

What gene therapy strategies show promise for TNNI3 mutations?

Several gene therapy approaches demonstrate potential for treating TNNI3-related cardiomyopathies:

  • Gene replacement/augmentation:

    • AAV-mediated delivery of wild-type TNNI3 has shown efficacy in preclinical models

    • Overexpression of wild-type TNNI3 ameliorates the RCM phenotype with the R170W mutation, suggesting a potential therapeutic target

    • Cardiotropic AAV serotypes (AAV9, AAV6) demonstrate efficient cardiac transduction

  • Gene editing approaches:

    • CRISPR-Cas9 correction of specific mutations

    • Base editing technologies for precise nucleotide changes

    • Prime editing for scarless genomic modification

  • RNA-based strategies:

    • Antisense oligonucleotides targeting mutant transcripts

    • siRNA-mediated selective knockdown of mutant alleles

    • RNA editing to correct mutations at the transcript level

  • Allele-specific silencing:

    • miRNA-based approaches targeting mutation-containing regions

    • Use of SNP-linked silencing strategies for dominant mutations

    • Targeted protein degradation of mutant TNNI3 protein

Research demonstrates that restoring adequate levels of functional TNNI3, particularly through overexpression of wild-type protein, can significantly improve relaxation impairment in cardiac tissues harboring TNNI3 mutations .

How can researchers effectively measure relaxation impairment in TNNI3 mutant heart tissue models?

Effective measurement of relaxation impairment requires:

  • Force measurement techniques:

    • High-resolution force transducers for isometric tension recording

    • Video-based motion tracking in EHT models

    • Atomic force microscopy for single-cell mechanics

  • Key parameters to assess:

    • Time to 50% and 90% relaxation (RT50, RT90)

    • Maximum relaxation velocity (dF/dt min)

    • Passive tension at various sarcomere lengths

    • Relaxation time constants (tau)

  • Calcium handling correlation:

    • Simultaneous calcium transient recording

    • Assessment of SERCA2a activity

    • Evaluation of NCX function

    • Measurement of calcium decay kinetics

  • Pharmacological interventions:

    • Response to beta-adrenergic stimulation

    • Force-frequency relationship assessment

    • Post-rest potentiation protocols

    • Response to calcium sensitizers/desensitizers

Engineered heart tissue constructs have proven particularly effective for detecting the impaired relaxation phenotype characteristic of TNNI3-related RCM, with greater sensitivity than traditional 2D culture systems .

What control mechanisms should be implemented when testing TNNI3 overexpression therapies?

Rigorous control mechanisms for TNNI3 overexpression studies include:

  • Genetic controls:

    • Empty vector delivery

    • Overexpression of irrelevant protein (e.g., GFP)

    • Expression of mutant TNNI3 (negative control)

    • Dose-response curve with varying expression levels

  • Temporal considerations:

    • Time-course evaluation post-intervention

    • Assessment of protein stability and turnover

    • Long-term follow-up for compensatory changes

  • Expression validation:

    • Western blot quantification of total TNNI3 levels

    • Distinction between endogenous and exogenous TNNI3

    • Assessment of isoform ratios (TNNI3:TNNI1)

    • Localization studies to confirm proper sarcomeric integration

  • Functional controls:

    • Wild-type cells treated identically

    • Rescue experiments in TNNI3-null backgrounds

    • Evaluation in multiple cell lines/genetic backgrounds

    • Comparison with pharmaceutical interventions

Research on TNNI3 overexpression has demonstrated improvement in relaxation parameters in engineered heart tissues harboring the R170W mutation, highlighting the importance of functional assessments in evaluating therapeutic efficacy .

How should researchers interpret contradictory calcium kinetics data in TNNI3 mutation studies?

When facing contradictory calcium kinetics data:

  • Methodological assessment:

    • Compare calcium dye properties (Fluo-4 vs. Fura-2 vs. genetically encoded indicators)

    • Evaluate recording conditions (temperature, stimulation protocol, solution composition)

    • Assess analysis algorithms and normalization methods

    • Consider temporal resolution limitations

  • Biological context evaluation:

    • Cell maturation status differences (TNNI1:TNNI3 ratio)

    • Subtype variations in cardiomyocyte populations (atrial vs. ventricular)

    • Compensatory changes in calcium handling proteins

    • Presence of non-myocytes affecting measurements

  • Integrated data analysis:

    • Correlate calcium measurements with contractile parameters

    • Evaluate consistency under varying loading conditions

    • Assess frequency-dependent behavior

    • Apply multiparametric statistical approaches

  • Validation strategies:

    • Reproduce findings using alternative technical approaches

    • Test in multiple biological replicates

    • Validate in different model systems

    • Compare with patient clinical data when available

Researchers should consider that calcium handling abnormalities in TNNI3 mutations may be secondary to altered myofilament properties rather than primary calcium cycling defects.

What bioinformatics tools are most useful for analyzing TNNI3 sequence variants and their potential pathogenicity?

For comprehensive TNNI3 variant analysis, researchers should utilize:

  • Variant annotation tools:

    • ANNOVAR for comprehensive annotation

    • VEP (Variant Effect Predictor) for functional impact prediction

    • MutationTaster for disease-causing potential assessment

    • CADD for integrative scoring of deleteriousness

  • Structural analysis software:

    • PyMOL for visualization of mutation sites within troponin complex

    • GROMACS for molecular dynamics simulations

    • FoldX for protein stability calculations

    • HADDOCK for protein-protein interaction modeling

  • Machine learning approaches:

    • CardioBoost for cardiac-specific variant interpretation

    • REVEL for ensemble prediction of pathogenicity

    • VEST for rare variant functional assessment

    • M-CAP for missense variant pathogenicity prediction

  • Population and clinical databases:

    • ClinVar for clinical significance assessment

    • gnomAD for population frequency data

    • HGMD for known disease-associated variants

    • ClinGen for expert-curated gene-disease relationships

Integration of multiple computational approaches with functional validation provides the most reliable assessment of TNNI3 variant pathogenicity.

How can researchers distinguish between primary effects of TNNI3 mutations and secondary adaptive responses?

Distinguishing primary from secondary effects requires:

  • Temporal analysis:

    • Implement inducible expression systems

    • Assess acute (hours to days) versus chronic (weeks) effects

    • Utilize time-series experiments with frequent sampling

    • Track the progression of phenotypic changes

  • Molecular pathway dissection:

    • Phosphoproteomic analysis to identify signaling cascades

    • Transcriptomic profiling at multiple timepoints

    • Targeted inhibition of key adaptive pathways

    • Combinatorial perturbation experiments

  • Systems biology approaches:

    • Network analysis to identify compensatory modules

    • Mathematical modeling of contractile dynamics

    • Principal component analysis of multi-omics data

    • Causal inference methodologies

  • Targeted intervention studies:

    • Rescue experiments with wild-type TNNI3

    • Pharmacological inhibition of specific signaling pathways

    • siRNA knockdown of putative mediators

    • Combined approach targeting multiple nodes

Primary effects typically manifest immediately following mutation introduction and directly involve troponin complex function, while secondary effects develop progressively and often involve transcriptional changes and protein remodeling.

What emerging technologies hold the most promise for advancing TNNI3 research?

Several cutting-edge technologies are poised to transform TNNI3 research:

  • Advanced cardiac tissue engineering:

    • Organ-on-chip platforms with multicellular compositions

    • Bioprinting technologies for precise spatial arrangement

    • Perfusable vascular networks in engineered tissues

    • Biomechanical stimulation systems mimicking hemodynamic forces

  • Novel genetic engineering approaches:

    • Prime editing for precise mutation correction

    • Base editing technologies for targeted nucleotide changes

    • CRISPR activation/inhibition for expression modulation

    • RNA editing tools for transcript modification

  • Single-cell technologies:

    • Single-cell proteomics for protein expression heterogeneity

    • Spatial transcriptomics for regional expression patterns

    • Single-cell contractility measurements

    • Multimodal omics integration at single-cell resolution

  • Advanced imaging modalities:

    • Super-resolution microscopy for sarcomeric organization

    • Label-free assessment of contractile dynamics

    • Intravital imaging of cardiac function in animal models

    • High-throughput automated phenotyping platforms

These technologies promise to provide unprecedented insights into the structural and functional consequences of TNNI3 mutations at molecular, cellular, tissue, and organ levels.

What are the key unresolved questions in TNNI3 research that warrant further investigation?

Critical unresolved questions include:

  • Mutation-specific mechanisms:

    • Why do some TNNI3 mutations cause HCM while others cause RCM?

    • How do mutations in different domains affect protein function?

    • What determines the age of onset and disease severity?

    • How do modifier genes influence phenotypic expression?

  • Therapeutic optimization:

    • What is the optimal dosage of wild-type TNNI3 for gene therapy?

    • Can allele-specific approaches effectively target dominant mutations?

    • What delivery methods provide the most efficient cardiac transduction?

    • How can off-target effects be minimized?

  • Developmental implications:

    • How do TNNI3 mutations affect cardiac development?

    • What is the relationship between TNNI1-to-TNNI3 switching and disease onset?

    • Do compensatory mechanisms differ between developing and mature hearts?

    • Can early intervention prevent disease progression?

  • Clinical translation:

    • What biomarkers predict disease progression in TNNI3 mutation carriers?

    • How can patient-specific therapeutic responses be predicted?

    • What is the optimal timing for therapeutic intervention?

    • How can engineered tissue models improve clinical decision-making?

Addressing these questions will require collaborative efforts across disciplines, combining basic science approaches with clinical investigations.

How might translational research bridge the gap between basic TNNI3 science and clinical applications?

Effective translational strategies include:

  • Improved disease modeling:

    • Patient-derived iPSC-CMs with enhanced maturation

    • EHT platforms calibrated to recapitulate clinical parameters

    • Humanized animal models for in vivo validation

    • Clinical tissue samples for direct comparison with models

  • Biomarker development:

    • Identification of circulating markers reflecting cardiac TNNI3 dysfunction

    • Imaging protocols specifically sensitive to diastolic abnormalities

    • Machine learning integration of multimodal data

    • Longitudinal studies correlating biomarkers with outcomes

  • Therapeutic pipeline optimization:

    • High-throughput screening platforms using EHT

    • Patient-specific response prediction algorithms

    • Combinatorial therapy approaches targeting multiple pathways

    • Delivery methods optimized for clinical translation

  • Clinical trial design:

    • Genetic stratification based on mutation characteristics

    • Surrogate endpoints reflecting early therapeutic efficacy

    • Adaptive trial designs for rare patient populations

    • Integration of digital health monitoring

Translational research must address the complete spectrum from molecular mechanisms to clinical outcomes, with EHT serving as a particularly valuable platform linking basic science discoveries to clinical applications in TNNI3-related cardiomyopathies .

Product Science Overview

Discovery and Importance

The discovery of troponin dates back to the early 1970s when researchers identified its role in muscle contraction. Troponin I, specifically, was found to inhibit the interaction between actin and myosin, thereby preventing muscle contraction in the absence of calcium ions . The cardiac-specific isoform of troponin I (cTnI) is unique to cardiac muscle cells and is not found in skeletal muscle, making it a highly specific biomarker for cardiac injury .

Structure and Function

Cardiac Troponin-I is a 24 kDa protein that binds to actin in thin myofilaments to hold the actin-tropomyosin complex in place. This binding prevents myosin from interacting with actin in relaxed muscle, thereby inhibiting contraction. Upon calcium binding to troponin C, a conformational change occurs, allowing the actin-myosin interaction and subsequent muscle contraction .

Clinical Significance

The clinical significance of cTnI lies in its role as a biomarker for myocardial infarction (heart attack). Elevated levels of cTnI in the blood are indicative of cardiac muscle damage. This makes cTnI an invaluable tool in the diagnosis and management of acute coronary syndromes (ACS). The development of highly sensitive assays for cTnI has further enhanced its utility in clinical practice .

Diagnostic Use

Cardiac Troponin-I levels are typically very low in healthy individuals, with the 99th percentile being less than a few nanograms per liter of blood. However, in the event of myocardial injury, cTnI levels rise significantly within a few hours, peaking at around 24 hours and remaining elevated for up to two weeks. This rapid and sustained increase in cTnI levels allows for the early detection and monitoring of myocardial infarction .

Advances in Assay Sensitivity

Recent advancements in assay sensitivity have led to the development of high-sensitivity cardiac troponin (hs-cTn) assays. These assays can detect even minor elevations in cTnI levels, allowing for the identification of subclinical myocardial injury. However, this increased sensitivity also means that cTnI can be elevated in conditions other than myocardial infarction, such as chronic kidney disease, heart failure, and extreme physical exertion .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.