TNNI3 Human, His

Cardiac Troponin-I Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Functional Role in Cardiac Muscle Physiology

Cardiac troponin I (cTnI) regulates actomyosin ATPase activity by modulating calcium sensitivity. It binds to troponin C and T, stabilizing the troponin-tropomyosin complex, which inhibits actin-myosin interaction at low calcium levels . Mutations in TNNI3 disrupt these interactions, leading to cardiomyopathies.

Post-Translational Modifications

cTnI undergoes phosphorylation (e.g., Ser23/Ser24 by PKA) and O-linked GlcNAcylation (Ser150), which modulate relaxation and contractility . These modifications are preserved in recombinant TNNI3 Human, His, enabling studies of their impact on cardiac function .

Research Applications and Disease Associations

Recombinant TNNI3 Human, His is used to study cardiomyopathy-associated mutations and develop diagnostic/prognostic biomarkers.

Hypertrophic Cardiomyopathy (HCM)

  • R192H Mutation: Increases calcium sensitivity and force generation in hiPSC-derived cardiomyocytes, mimicking HCM pathology .

  • I195F Variant: Linked to pediatric HCM with arrhythmias and myocardial fibrosis .

Dilated Cardiomyopathy (DCM)

  • K36Q Mutation: Reduces force generation and calcium sensitivity, causing early-onset DCM .

  • Homozygous Null Mutations: Lead to severe neonatal DCM with left ventricular dilation .

Restrictive Cardiomyopathy and Others

  • TNNI3 Mutations: Impair actin binding, causing diastolic dysfunction .

  • Left Ventricular Noncompaction (LVNC): Rarely linked to TNNI3 variants (e.g., R192H) .

Allele Frequencies in HCM Cohorts

VariantPopulationHCM Cases (AF)Controls (AF)Odds Ratio
TNNI3:p.R79CSingaporean Chinese0.017860.005503.33
TNNI3:p.R286HSingaporean Chinese0.022320.0016514.1

AF = allele frequency; Data from Singaporean HCM studies .

Functional Studies in hiPSC-Derived Cardiomyocytes

High-throughput assays using TNNI3 Human, His variants reveal:

VariantPhenotypeMechanism
R192H (HCM)↑ Peak force, diastolic tension, calcium sensitivity Enhanced calcium binding affinity
K36Q (DCM)↓ Peak force, calcium sensitivity Impaired actin-troponin interaction

These models validate TNNI3 mutations as disease drivers and support personalized medicine approaches .

Diagnostic and Prognostic Biomarker Potential

Recombinant TNNI3 Human, His is used in ELISA assays to detect circulating cTnI levels, though native protein studies are prioritized for clinical relevance .

Challenges and Future Directions

  • Penetrance Variability: TNNI3 mutations show low penetrance (~50%), complicating risk stratification .

  • Therapeutic Targets: Modulating cTnI phosphorylation or calcium binding may offer novel therapies .

Product Specs

Introduction
Troponin I (TnI), along with troponin T (TnT) and troponin C (TnC), constitute the troponin complex found in the thin filaments of striated muscle. TnI plays a crucial role as the inhibitory subunit, effectively preventing actin-myosin interactions and thereby regulating striated muscle relaxation. The TnI subfamily comprises three genes: TnI-skeletal-fast-twitch, TnI-skeletal-slow-twitch, and TnI-cardiac. Notably, the TNNI3 gene is responsible for encoding the TnI-cardiac protein and exhibits exclusive expression within cardiac muscle tissues. Mutations in the TNNI3 gene have been implicated in the development of familial hypertrophic cardiomyopathy type 7 (CMH7) and familial restrictive cardiomyopathy (RCM).
Description
Recombinant Human TNNI3 protein, expressed in E. coli, is a full-length protein encompassing 260 amino acids. A 6-His tag is fused at the C-terminus, resulting in a protein that migrates as a 27 kDa band on SDS-PAGE. The protein has been purified using a proprietary chromatographic technique.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
TNNI3 protein is supplied in a solution containing PBS and 25mM arginine.
Purity
The purity of the protein exceeds 95%, as determined by 12% SDS-PAGE analysis with Coomassie Blue staining.
Stability
For optimal long-term storage, TNNI3 should be stored at a temperature below -18°C. While the protein remains stable at 4°C for a period of one week, repeated freeze-thaw cycles should be avoided.
Synonyms
Troponin I cardiac muscle, Cardiac troponin I, TNNI3, TNNC1, CMH7, RCM1, cTnI, CMD2A, MGC116817.
Source
Escherichia Coli.

Q&A

What is TNNI3 and what role does it play in cardiac function?

TNNI3, also known as cardiac troponin I, is a thin filament protein essential for calcium-mediated regulation of cardiac muscle contraction. It functions as part of the troponin complex that includes troponin T and troponin C. When calcium binds to troponin C, it induces a conformational change in troponin I that allows myosin-actin interaction and subsequent muscle contraction. TNNI3 is exclusively expressed in cardiac tissue, making it a cardiac-specific marker. Mutations in the TNNI3 gene can disrupt calcium cycling and contractile function, leading to various forms of cardiomyopathy . Research methodologically distinguishes TNNI3 from its skeletal muscle counterparts (TNNI1 and TNNI2) based on its unique protein sequence and tissue-specific expression patterns.

How prevalent are TNNI3 mutations in different types of cardiomyopathies?

TNNI3 mutations account for approximately 2-7% of familial cardiomyopathy cases, representing a relatively small but significant proportion of genetic causes . In the literature, the majority of TNNI3 mutations (approximately 85%) are associated with hypertrophic cardiomyopathy (HCM), followed by restrictive cardiomyopathy (RCM) at around 8%, while dilated cardiomyopathy (DCM) cases are rare . This distribution pattern has been consistently observed across multiple population studies. The Dutch population study reported 14 different mutations in 30 families, with certain mutations (p.Arg145Trp, p.Ser166Phe, and p.Glu209Ala) occurring at relatively higher frequencies . The relatively low prevalence might be explained by the severe clinical outcomes associated with certain mutations, potentially reducing reproductive fitness in affected individuals.

What is the clinical presentation of TNNI3 mutation carriers?

TNNI3 mutations typically cause early-onset cardiomyopathy with severe clinical manifestations. The mean age at diagnosis is 36.5 years, though presentation can range from infancy to late adulthood . Clinical heterogeneity is substantial, with phenotypes including:

  • Hypertrophic cardiomyopathy (HCM): Characterized by left ventricular hypertrophy, often with septum involvement (interventricular septum thickness ranging from 14-30mm)

  • Restrictive cardiomyopathy (RCM): Characterized by impaired ventricular filling with normal wall thickness and systolic function

  • Dilated cardiomyopathy (DCM): Less common, characterized by ventricular dilation and reduced contractile function

Notably, even within families carrying the same mutation, different phenotypes can be observed, suggesting the influence of modifier genes or environmental factors . RCM-causing mutations tend to manifest in very young children and generally carry a poorer prognosis. Some mutations, such as the founder mutation p.Arg145Trp, display particularly variable expressivity, ranging from asymptomatic carriers to individuals with severe disease and sudden cardiac death .

How can I establish an effective hiPSC-CM model for studying TNNI3 variants?

Establishing an effective hiPSC-CM model for TNNI3 variant studies requires addressing the challenge that hiPSC-CMs predominantly express TNNI1 rather than TNNI3, complicating the assessment of TNNI3 variant effects. A methodologically sound approach involves:

  • Generation of TNNI1-/-TNNI3-/- double knockout hiPSCs using CRISPR-Cas9 technology

  • Differentiation of these cells into cardiomyocytes following standard cardiac differentiation protocols

  • Lentiviral transduction of wild-type or mutant TNNI3 during replating onto appropriate substrates (e.g., polyacrylamide gels with fluorescent beads for contractility tracking)

  • Incubation for 5-7 days to allow for stable expression

  • Functional assessment using calcium-sensitive fluorescent dyes combined with high-throughput imaging platforms

This approach allows for precise control of TNNI3 expression and direct comparison between wild-type and variant forms without interference from endogenous troponin expression. The double knockout strategy is crucial, as it eliminates compensation by TNNI1, which would otherwise mask the effects of TNNI3 variants.

What parameters should be measured when characterizing TNNI3 variants in cellular models?

Comprehensive characterization of TNNI3 variants should include multiple functional parameters that reflect the physiological roles of cardiac troponin I:

  • Contractility measurements:

    • Peak force generation

    • Diastolic tension

    • Contractile kinetics (time to peak, relaxation time)

  • Calcium handling parameters:

    • Calcium sensitivity (force-calcium relationship)

    • Calcium transient amplitude and kinetics

    • Diastolic calcium levels

  • Sarcomeric organization:

    • Myofilament structure and organization

    • Protein-protein interactions within the troponin complex

Research has demonstrated that HCM-associated variants (e.g., R192H) typically show increased peak force, elevated diastolic tension, and heightened calcium sensitivity, while DCM-associated variants (e.g., K36Q) typically exhibit decreased peak force and reduced calcium sensitivity compared to wild-type TNNI3 . These distinct functional signatures provide valuable insights into pathogenic mechanisms and correlate with clinical phenotypes.

How can ELISA be optimized for quantifying TNNI3 in research samples?

Optimizing ELISA for TNNI3 quantification requires careful attention to several methodological considerations:

  • Antibody selection: Use a solid-phase sandwich ELISA with matched antibody pairs that specifically recognize both natural and recombinant human TNNI3. The capture antibody should be pre-coated in microplate wells, with a detector antibody that recognizes a different epitope .

  • Sample preparation: For serum/plasma samples, standardize collection timing, particularly for studies examining cardiac injury where TNNI3 levels may fluctuate. For cell culture studies, optimize protein extraction protocols to ensure complete solubilization of TNNI3 from myofilaments.

  • Standard curve generation: Use purified recombinant TNNI3 with confirmed purity and concentration for accurate quantification. Generate a standard curve covering the physiological and pathological range expected in your experimental samples.

  • Validation steps:

    • Confirm specificity by testing cross-reactivity with skeletal troponin isoforms

    • Determine precision through intra- and inter-assay coefficient of variation assessment

    • Verify linearity of dilution across the working range

    • Establish recovery rates using spike-in controls

The intensity of signal generated should be directly proportional to the concentration of TNNI3 present in the original specimen, allowing for accurate quantification across experimental conditions .

What genotype-phenotype correlations have been established for TNNI3 mutations?

Several important genotype-phenotype correlations have been established for TNNI3 mutations through comprehensive genetic and clinical studies:

MutationPredominant PhenotypeAge of OnsetClinical SeverityKnown Mechanisms
p.Arg145TrpHCM/RCMVariable (9-47 years)Severe, SCD riskIncreased Ca2+ sensitivity
p.Arg145GlnRCM<9 yearsSevereIncreased Ca2+ sensitivity
p.Ala157ValHCM26-46 yearsModerate-severeAltered thin filament regulation
p.Ser166PheHCM39-58 yearsVariableFounder mutation in Netherlands
p.Lys178delRCM<11 yearsSevereDe novo mutation
p.Glu209AlaHCM27-62 yearsVariableNon-founder recurring mutation

These correlations reveal several patterns:

  • Mutations affecting the inhibitory region of TNNI3 (e.g., arginine 145) tend to cause more severe phenotypes with earlier onset

  • RCM-causing mutations generally present in childhood with poor prognosis

  • HCM-causing mutations show variable expressivity, even within families

  • Certain positions (like 145, 157, and 209) appear to be mutational hotspots

Notably, some mutations demonstrate remarkable clinical heterogeneity. For example, p.Arg145Trp can cause both HCM and RCM phenotypes even within the same family, suggesting the presence of genetic modifiers or environmental factors influencing disease expression .

How do founder mutations contribute to our understanding of TNNI3 pathogenicity?

Founder mutations provide valuable insights into disease mechanisms and population genetics of TNNI3-related cardiomyopathies. In the Dutch population, haplotype analysis confirmed that p.Arg145Trp and p.Ser166Phe are founder mutations, while p.Glu209Ala, despite its recurrence, is not . These findings have several important implications:

  • Clinical variability: Founder mutations demonstrate wide phenotypic variation despite identical genetic defects. The p.Arg145Trp mutation, the most frequent TNNI3 mutation in the Netherlands (27% of cases), shows variable disease expression and age of onset, even within families . This variability suggests that additional genetic or environmental factors significantly modify disease expression.

  • Penetrance patterns: Founder mutations allow assessment of penetrance across multiple unrelated families. For p.Arg145Trp carriers, disease onset ranged from before age 9 to adulthood, with some carriers developing RCM while others presented with HCM .

  • Geographic distribution: The random distribution of founder mutations across the Netherlands suggests these mutations originated many generations ago and have spread throughout the population .

  • Modifier gene hypothesis: The variable expressivity of founder mutations strongly supports the existence of modifier genes that influence disease phenotype. These could include other sarcomeric genes, calcium handling proteins, or genes involved in cardiac energy metabolism.

Understanding founder mutations helps distinguish between pathogenic variants and benign polymorphisms, particularly for variants of uncertain significance, by providing population context for genetic findings.

What are the most effective approaches for functional characterization of TNNI3 variants?

The most effective functional characterization of TNNI3 variants employs a multi-level experimental approach:

  • Cellular models:

    • hiPSC-CMs with TNNI1/TNNI3 double knockout background for clean genetic context

    • Lentiviral transduction of wild-type or mutant TNNI3

    • Measurement of contractility, calcium sensitivity, and sarcomere function

  • Biochemical characterization:

    • Protein-protein interaction studies with other troponin components

    • Phosphorylation analyses to assess post-translational regulation

    • Structural studies (e.g., crystallography, cryo-EM) to determine effects on protein conformation

  • Tissue-level studies:

    • Engineered heart tissues from hiPSC-CMs expressing TNNI3 variants

    • Ex-vivo analysis of heart tissue from animal models or patient samples

    • Force-calcium relationship measurements in permeabilized myocardial strips

  • In vivo models:

    • Transgenic mouse models expressing human TNNI3 variants

    • Long-term phenotypic assessment including echocardiography, ECG, and histology

How can contradictions in TNNI3 functional data be resolved?

Resolving contradictions in TNNI3 functional data requires systematic analysis of experimental variables and methodological differences:

  • Model system considerations:

    • Different model systems (bacterial expression, insect cells, mammalian cells) may yield conflicting results due to differences in post-translational modifications and protein folding

    • Expression of TNNI3 in isolation versus within the troponin complex can significantly impact findings

    • The presence of endogenous troponin isoforms in hiPSC-CMs can mask effects of TNNI3 variants

  • Experimental conditions:

    • Temperature, pH, and ionic strength significantly affect troponin function

    • Calcium concentration ranges must be physiologically relevant

    • Loading conditions for contractility measurements must be standardized

  • Methodological approach:

    • Implement double knockout strategies to eliminate interference from endogenous troponins

    • Use multiple complementary techniques to assess the same parameter

    • Include appropriate controls (wild-type, known pathogenic variants, benign variants)

  • Data integration strategies:

    • Develop standardized scoring systems to classify variants based on multiple functional parameters

    • Consider allele-specific effects and dose-dependency

    • Correlate functional findings with clinical data from carriers of the same variant

When faced with contradictory data, researchers should prioritize findings from systems that most closely recapitulate human cardiac physiology and validate results across multiple experimental platforms.

What controls are essential for validating TNNI3 functional assays?

Rigorous validation of TNNI3 functional assays requires inclusion of multiple control types:

  • Positive and negative controls:

    • Wild-type TNNI3 as baseline reference

    • Well-characterized pathogenic variants (e.g., R192H for HCM, K36Q for DCM)

    • Known benign variants/polymorphisms

    • Empty vector controls to assess effects of viral transduction

  • Methodological controls:

    • Confirmation of knockout efficiency in TNNI1-/-TNNI3-/- models

    • Verification of protein expression levels for all TNNI3 variants tested

    • Assessment of cell viability and maturity across experimental conditions

  • Physiological controls:

    • Response to β-adrenergic stimulation to confirm physiological regulation

    • Demonstration of Frank-Starling relationship (length-dependent activation)

    • Calcium dose-response curves covering physiological range

  • Internal validation controls:

    • Technical replicates to assess precision

    • Biological replicates from independent differentiations/transductions

    • Time-course studies to ensure stability of the phenotype

How can TNNI3 genetic testing results be integrated into clinical management?

Integrating TNNI3 genetic testing results into clinical management requires a comprehensive approach:

  • Risk stratification based on variant-specific data:

    • TNNI3 mutations are associated with early onset and severe clinical presentation

    • RCM-causing mutations, particularly those presenting in childhood, warrant close monitoring due to poor prognosis

    • Certain mutations (e.g., p.Arg145Trp) have been associated with sudden cardiac death risk

  • Family screening considerations:

    • Cascade genetic testing should be offered to first-degree relatives

    • Clinical evaluation should be tailored based on the specific mutation identified

    • Variable expressivity means that even asymptomatic carriers require regular cardiac evaluation

  • Treatment implications:

    • Early identification of high-risk mutations may influence timing of interventions

    • Understanding the molecular mechanism (increased vs. decreased calcium sensitivity) may guide precision medicine approaches

    • Emerging targeted therapies addressing specific contractile abnormalities may be particularly relevant for TNNI3 mutation carriers

  • Surveillance protocols:

    • Frequency of cardiac imaging may be determined by mutation-specific risk profiles

    • ECG, echocardiography, and cardiac MRI should be considered in comprehensive evaluation

    • Consideration of implantable cardioverter-defibrillators for high-risk mutations with sudden death history

The Dutch population study demonstrated that some TNNI3 mutations (like p.Arg145Trp) can cause variable disease expression within families, highlighting the need for individualized assessment rather than mutation-based protocols alone .

What are the current therapeutic approaches targeting TNNI3 dysfunction?

Current therapeutic approaches targeting TNNI3 dysfunction are advancing on multiple fronts:

  • Small molecule modulators:

    • Calcium sensitizers (for DCM-associated variants) that enhance cardiac contractility

    • Calcium desensitizers (for HCM-associated variants) that normalize increased calcium sensitivity

    • Post-translational modification regulators that target phosphorylation status of TNNI3

  • Gene-based therapies:

    • Allele-specific silencing to reduce expression of mutant TNNI3

    • CRISPR-based approaches for gene correction in patient-derived cells

    • Gene replacement strategies delivering wild-type TNNI3

  • Protein-based interventions:

    • Engineered troponin complexes that compete with mutant proteins

    • Peptides targeting specific interaction domains of TNNI3

    • Stabilizers of proper troponin complex formation

  • Precision medicine strategies:

    • Functional characterization of patient-specific variants to guide medication selection

    • Computational modeling to predict therapeutic responses based on structural effects

    • hiPSC-CM drug screening platforms for personalized therapy selection

These approaches are informed by mechanistic research demonstrating that HCM-associated variants typically increase calcium sensitivity while DCM-associated variants decrease it , providing a rational basis for therapeutic targeting of these specific functional abnormalities.

What emerging technologies will advance TNNI3 research?

Several emerging technologies hold promise for advancing TNNI3 research:

  • Advanced cellular models:

    • Engineered 3D cardiac tissues that better recapitulate adult cardiac physiology

    • Microfluidic "heart-on-chip" platforms for high-throughput functional screening

    • Organoid models incorporating multiple cell types for studying cell-cell interactions

  • Next-generation genetic approaches:

    • Base editing and prime editing for precise correction of TNNI3 mutations

    • Single-cell transcriptomics to understand cellular heterogeneity in response to TNNI3 mutations

    • Long-read sequencing to detect complex structural variants affecting TNNI3 regulation

  • Imaging innovations:

    • Super-resolution microscopy for visualization of sarcomeric protein interactions

    • Label-free imaging techniques for non-invasive assessment of contractile function

    • Intravital imaging in animal models for real-time monitoring of cardiac function

  • Computational approaches:

    • Machine learning algorithms for predicting variant pathogenicity

    • Molecular dynamics simulations to model TNNI3 conformational changes

    • Systems biology approaches to integrate multi-omics data

The high-throughput functional determination of TNNI3 variants using gene replacement and physiologic imaging platforms represents a significant methodological advance that will enable comprehensive characterization of the numerous TNNI3 variants currently classified as of uncertain significance.

How can large-scale collaborative efforts enhance our understanding of TNNI3 variants?

Large-scale collaborative efforts can substantially advance TNNI3 research through:

  • Population-level variant analysis:

    • Pooling genetic data across diverse populations to identify novel variants

    • Establishing accurate prevalence rates for known pathogenic variants

    • Identifying founder mutations and their geographic distribution

  • Standardized functional assessment:

    • Development of consensus protocols for functional characterization

    • Creation of reference datasets for known pathogenic and benign variants

    • Establishment of centralized repositories for functional data sharing

  • Clinical data integration:

    • Creation of detailed phenotype-genotype databases linking TNNI3 variants to clinical outcomes

    • Longitudinal follow-up studies to assess disease progression across large cohorts

    • Meta-analyses of treatment responses in carriers of specific mutations

  • Interdisciplinary approaches:

    • Collaboration between basic scientists, clinical cardiologists, geneticists, and bioinformaticians

    • Integration of structural biology, functional studies, and clinical observations

    • Development of translational pipelines from variant discovery to therapeutic testing

The Dutch cardiomyopathy study demonstrates the value of multi-center collaboration, combining patients and genetic data from four cardiogenetic centers to achieve a comprehensive overview of TNNI3 mutations in a national population . Similar collaborative efforts at international scales would provide even greater insights into the spectrum of TNNI3 variants and their clinical significance.

Product Science Overview

Introduction

Cardiac Troponin-I (cTnI) is a critical protein in the regulation of cardiac muscle contraction. It is part of the troponin complex, which includes Troponin T (TnT) and Troponin C (TnC). This complex plays a pivotal role in the calcium-mediated regulation of muscle contraction in the heart. The recombinant form of Cardiac Troponin-I, tagged with a histidine (His) sequence, is widely used in research and diagnostic applications.

Structure and Function

Cardiac Troponin-I is a 209 amino acid protein with a molecular weight of approximately 23.9 kDa . It is unique among the troponins due to its N-terminal extension of 26 amino acids . This protein is specifically expressed in cardiac muscle tissue and is involved in inhibiting the actomyosin ATPase activity in the absence of calcium, thereby preventing muscle contraction.

The His tag, a sequence of histidine residues, is added to the recombinant protein to facilitate its purification and detection. This tag binds strongly to nickel ions, allowing for easy separation of the recombinant protein from other cellular components using affinity chromatography.

Expression and Purification

Recombinant Cardiac Troponin-I is typically expressed in Escherichia coli (E. coli) systems . The protein is then purified using chromatographic techniques, often involving nickel-affinity columns due to the His tag. The resulting product is highly pure, with a purity level exceeding 90% .

Applications
  1. Diagnostic Marker: Cardiac Troponin-I is a highly sensitive and specific marker for myocardial damage. Elevated levels of cTnI in the serum are indicative of myocardial infarction (heart attack) or other forms of cardiac injury .
  2. Research: The recombinant form of cTnI is used in various research applications to study cardiac muscle physiology, the mechanisms of muscle contraction, and the effects of different drugs on cardiac function .
  3. Drug Development: It is also used in high-throughput screening assays to identify potential therapeutic compounds that can modulate cardiac muscle contraction .
Stability and Storage

Recombinant Cardiac Troponin-I is stable for up to six months when stored at temperatures lower than -70°C . Upon reconstitution, it remains stable for up to one week at 4°C or up to three months at -20°C. For long-term storage, it is recommended to add a carrier protein, such as 0.1% Bovine Serum Albumin (BSA), to prevent degradation .

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.