TNNT2 Human

Cardiac Muscle Troponin T Recombinant Human
Shipped with Ice Packs
In Stock

Description

Introduction and Overview

The TNNT2 gene encodes cardiac troponin T (cTnT), a critical subunit of the troponin complex in cardiac muscle. This protein regulates muscle contraction by modulating calcium-dependent interactions between actin and myosin filaments. Mutations in TNNT2 are linked to hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and restrictive cardiomyopathy (RCM), underscoring its vital role in cardiac function .

Troponin Complex Dynamics

The troponin complex (cTnT, TnI, TnC) anchors to the thin filament and regulates contraction:

  1. Low calcium: cTnT binds tropomyosin, blocking actomyosin interaction .

  2. High calcium: Structural changes in cTnT enable tropomyosin displacement, permitting contraction .

Developmental Regulation

  • Embryonic heart: Exon 5 inclusion increases negative charge, enhancing calcium sensitivity and force production .

  • Adult heart: Exon 5 exclusion reduces calcium sensitivity, optimizing energy efficiency .

Cardiomyopathies Linked to TNNT2

CardiomyopathyMutation EffectsExample Mutations
HCMHypercontractility, increased calcium affinityR92Q, E160Δ, R134G
DCMHypocontractility, decreased calcium affinityK210Δ, R278C
RCMStiff sarcomeres, restrictive physiologyΔE96, E136K

Key Findings:

  • HCM vs. DCM: HCM mutations increase microtissue contraction (e.g., R92Q), while DCM mutations reduce it (e.g., K210Δ) .

  • Variable expressivity: Same TNNT2 mutation (e.g., p.Arg92Gln) causes severe HCM in one family member and subclinical symptoms in another .

Functional Genomics Platform

A study using human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes evaluated 51 TNNT2 variants :

  1. Sarcomere Function:

    • HCM variants → Increased cardiac microtissue contraction.

    • DCM variants → Decreased contraction.

  2. Transcriptomic Changes:

    • 101 gene transcripts altered, including NPPB (atrial natriuretic peptide) .

  3. Variant Reclassification:

    • 2 pathogenic/likely pathogenic variants reclassified as benign.

    • 2 variants of uncertain significance flagged as pathogenic .

Diagnostic Tools

  • NPPB reporter assay: Measures NPPB expression to predict variant pathogenicity .

  • Calcium affinity assays: Confirm altered myofilament calcium transients in HCM/DCM variants .

Evolutionary Conservation

  • TNNT2 is conserved across vertebrates, paired with slow skeletal TnI in embryonic heart .

  • Alternative splicing in TNNT2 is regulated by a "biological clock," synchronizing exon inclusion across species .

Alternative Splicing in Development

ExonFunction
4Minor regulatory role, exact function unknown
5Embryonic expression → Higher calcium sensitivity
13Between middle/C-terminal regions → Unknown functional impact

Product Specs

Introduction
TNNT2 is the tropomyosin-binding subunit of the troponin complex, which is located on the thin filament of striated muscles. It regulates muscle contraction in response to changes in intracellular calcium ion concentration. Mutations in the TNNT2 gene can cause familial hypertrophic cardiomyopathy and dilated cardiomyopathy. TNNT2 mutation is implicated in hypertrophic cardiomyopathy.
Description
Recombinant Human Cardiac Muscle Troponin T, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 297 amino acids with a molecular weight of 35.8 kDa. TNNT2 is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile filtered colorless solution.
Formulation
TNNT2 is supplied as a 0.2 µm filtered solution containing 10 mM Tris-HCl (pH 8.0), 50% glycerol, and 250 mM NaCl.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 98.0% as determined by: (a) RP-HPLC analysis and (b) SDS-PAGE analysis.
Synonyms

CMH2, RCM3, TnTC, cTnT, CMPD2, MGC3889, TNNT2, Troponin T- cardiac muscle, Cardiac muscle troponin T, troponin T type 2 (cardiac).

Source

Escherichia Coli.

Amino Acid Sequence

SDIEEVVEEY EEEEQEEAAV EEEEDWREDE DEQEEAAEED AEAEAETEET RAEEDEEEEE AKEAEDGPME ESKPKPRSFM PNLVPPKIPD GERVDFDDIH RKRMEKDLNE LQALIEAHFE NRKKEEEELV SLKDRIERRR AERAEQQRIR NEREKERQNR LAEERARREE EENRRKAEDE ARKKKALSNM MHFGGYIQKQ AQTERKSGKR QTEREKKKKI LAERRKVLAI DHLNEDQLRE KAKELWQSIY NLEAEKFDLQ EKFKQQKYEI NVLRNRINDN QKVSKTRGKA KVTGRWK.

Q&A

What is TNNT2 and what is its fundamental role in cardiac function?

TNNT2 encodes cardiac troponin T (cTnT), a critical component of the troponin complex in cardiac sarcomeres. It functions as the structural and regulatory link between the troponin complex and tropomyosin, playing a vital role in calcium-dependent regulation of cardiac muscle contraction. The protein mediates the interaction between calcium binding to troponin C and the subsequent movement of tropomyosin, allowing actin-myosin cross-bridge formation essential for cardiac contractility .

How can researchers distinguish between different functional domains of the TNNT2 protein?

TNNT2 contains multiple functional domains that can be experimentally characterized using domain-specific antibodies, recombinant protein assays, and targeted mutagenesis approaches. Key regions include the N-terminal tropomyosin-binding domain, central regions interacting with troponin C and troponin I, and the C-terminal domain that stabilizes the troponin complex. Understanding these domains is essential when interpreting variant locations, as mutations in different regions produce distinct functional consequences and can affect experimental readouts differently .

What distinguishes hypertrophic cardiomyopathy (HCM) from dilated cardiomyopathy (DCM) TNNT2 variants at the functional level?

HCM-associated and DCM-associated TNNT2 variants demonstrate opposite functional effects in experimental models:

PhenotypeContractilityMyofilament Ca²⁺ SensitivityNPPB ExpressionExample Variants
HCMIncreasedIncreasedElevatedR92Q, E160Δ, I79N
DCMDecreasedDecreasedReducedR134G, K210Δ, R141W
These contrasting functional profiles support a sarcomere tension-based model of cardiomyopathy, where the direction of change in contractile function determines the disease phenotype, regardless of the variant's specific location within the protein .

How can researchers experimentally determine the pathogenicity of TNNT2 variants of uncertain significance?

Determining pathogenicity requires a multi-modal experimental approach:

  • Establish isogenic cell lines using CRISPR/Cas9 genome editing to introduce the variant of interest

  • Perform cardiac microtissue (CMT) force measurements to assess contractility changes

  • Utilize transcriptomic analysis (RNA-seq) to identify altered gene expression patterns

  • Implement reporter assays such as NPPB→tdTomato to monitor sarcomere dysfunction

  • Measure myofilament-directed calcium transients to assess calcium sensitivity alterations
    This integrated approach has successfully classified 51 TNNT2 variants, including reclassification of several variants of uncertain significance based on functional evidence .

What limitations should researchers consider when interpreting population frequency data for TNNT2 variants?

Population frequency data alone has limited utility for TNNT2 variant classification. Research shows that while variants with >1 allele count in population databases like ExAC typically demonstrate benign-like functional characteristics, many extremely rare variants (0-1 allele count) also show wild-type-like function. In a comprehensive analysis of 51 TNNT2 variants, approximately 15.6% of variants with 0 allele counts in ExAC functionally resembled wild-type TNNT2, highlighting that rarity alone is insufficient for predicting pathogenicity . Researchers should therefore combine population data with functional assays for more accurate classification.

What are the key considerations when designing human induced pluripotent stem cell (hiPSC) models for TNNT2 variant studies?

When designing hiPSC models for TNNT2 research, researchers should consider:

  • Genetic background selection: Use well-characterized lines or patient-derived lines relevant to the research question

  • Variant introduction strategy: Choose between isogenic models (CRISPR/Cas9 editing) or transgenic approaches (like SarcTg lentiviral system)

  • Expression levels: Ensure physiological-like cTnT expression levels to avoid artifacts from overexpression

  • Differentiation protocol: Optimize cardiac differentiation for consistent sarcomere assembly and maturation

  • Controls: Include both wild-type controls and established pathogenic variants (e.g., R92Q for HCM, R134G for DCM) as benchmarks

  • Functional readouts: Implement multiple assays at different scales (molecular, cellular, tissue) for comprehensive characterization

What methodological approaches can mitigate the limitations of hiPSC-derived cardiomyocytes in TNNT2 research?

hiPSC-derived cardiomyocytes (hiPSC-CMs) have several limitations, including their immature phenotype resembling neonatal rather than adult cardiomyocytes. To address these limitations:

  • Implement extended culture periods (>60 days) to promote maturation

  • Use mechanical or electrical stimulation to enhance structural and functional maturation

  • Employ 3D culture systems like cardiac microtissues to better recapitulate tissue architecture

  • Consider the expression of fetal isoforms (e.g., TNNI1 instead of TNNI3) when interpreting calcium sensitivity results

  • Benchmark transgenic models against isogenic models for established mutations to validate findings

  • Complement hiPSC-CM studies with other model systems when appropriate (e.g., animal models, recombinant protein assays)

  • Account for cell line variability by using multiple lines or isogenic controls

How do transgenic models compare to isogenic models in TNNT2 functional studies?

AspectTransgenic (SarcTg) ApproachIsogenic CRISPR/Cas9 Models
EfficiencyHigh-throughput screening of multiple variantsTime-intensive generation of individual lines
Genetic contextCo-expression with endogenous WT proteinHeterozygous expression mimicking patient genotype
Expression controlPotential variability in expression levelsPhysiological expression from endogenous locus
Background effectsCan study variants in WT or knockout backgroundDirect comparison in identical genetic background
ValidationRequires benchmarking against established variantsGold standard for variant characterization
ScalabilitySuitable for screening large variant catalogsLimited by time and resource constraints
Research demonstrates that for well-established TNNT2 variants like R92Q (HCM) and R134G (DCM), both approaches show concordant contractility phenotypes, supporting the validity of the transgenic approach for large-scale variant screening .

How can researchers leverage transcriptomic data to understand downstream effects of TNNT2 variants?

RNA sequencing analysis reveals that sarcomere contractile function exhibits graded regulation of 101 gene transcripts in response to TNNT2 variants. Researchers can:

  • Perform differential expression analysis between variant and wild-type samples

  • Conduct pathway enrichment analysis using tools like MSigDB to identify affected pathways

  • Focus on established biomarkers like NPPB, which shows strong correlation with sarcomere function

  • Analyze MAPK signaling targets and cardiac-specific transcription factors like HOPX

  • Develop transcriptional reporters (e.g., NPPB→tdTomato) based on identified transcripts to monitor variant effects

  • Use transcriptomic signatures to distinguish between HCM-like and DCM-like functional impacts

  • Correlate transcriptomic changes with contractile phenotypes to establish mechanistic links
    This approach not only helps classify variants but also provides insights into the molecular mechanisms of disease pathogenesis.

What is the SarcTg platform and how can it be implemented for high-throughput TNNT2 variant screening?

The SarcTg (Sarcomere Transgenic) platform is a comprehensive functional genomics approach for TNNT2 variant characterization that includes:

  • Genetic components:

    • CRISPR/Cas9-engineered TNNT2 knockout (SarcKO) hiPSC line

    • Lentiviral delivery system for variant expression

    • NPPB→tdTomato reporter integration at the endogenous NPPB locus

  • Implementation protocol:

    • Generate lentiviruses encoding TNNT2 variants of interest

    • Transduce the viruses into NPPB→tdTomato SarcKO hiPSC-CMs

    • Differentiate cells into cardiomyocytes following standard protocols

    • Quantify tdTomato intensity by flow cytometry relative to wild-type controls

    • Include R92Q (HCM) and R134G (DCM) controls in each experiment for validation

  • Data analysis:

    • Compare variant-induced reporter activity to wild-type TNNT2

    • Classify variants based on significant deviations in reporter signal

    • Confirm selected variants using cardiac microtissue contractility assays
      This platform successfully discriminated 28 of 30 pathogenic/likely pathogenic variants from wild-type controls with 93.3% accuracy.

How can researchers assess calcium sensitivity changes in TNNT2 variants?

To assess altered calcium handling in TNNT2 variants, researchers can implement a thin filament–directed calcium reporter system that specifically monitors myofilament calcium affinity:

  • Assay setup:

    • Express calcium sensors localized to thin filaments in hiPSC-CMs

    • Introduce TNNT2 variants via lentiviral transduction or genome editing

    • Measure calcium transients during spontaneous or paced contractions

    • Compare thin filament calcium signals between variant and wild-type samples

  • Key findings:

    • HCM-associated variants (R92Q, E160Δ) increased myofilament-directed calcium transients

    • DCM-associated variants (R134G, K210Δ) decreased these transients

    • Effects occurred regardless of proximity to troponin C binding sites, suggesting allosteric mechanisms

  • Interpretation:

    • Changes in thin filament calcium sensitivity directly correlate with contractile phenotypes

    • This mechanism appears to be a fundamental driver of pathogenicity rather than a secondary effect
      Understanding these changes provides crucial insights into variant pathophysiology and potential therapeutic targets.

What methodological approaches can quantify the contractile effects of TNNT2 variants?

Cardiac microtissue (CMT) force measurements provide direct quantification of TNNT2 variant effects on contractility:

  • Experimental setup:

    • Generate 3D engineered cardiac tissues using hiPSC-CMs expressing TNNT2 variants

    • Mount tissues between flexible pillars or force transducers

    • Record spontaneous or electrically stimulated contractions

    • Analyze twitch force, kinetics, and frequency dependence

  • Parameters to measure:

    • Maximum twitch force (amplitude)

    • Time to peak contraction

    • Relaxation kinetics (T50, time to 50% relaxation)

    • Force-frequency relationship

    • Response to β-adrenergic stimulation

  • Validation approaches:

    • Compare transgenic variant expression to isogenic models

    • Correlate force measurements with calcium sensitivity data

    • Link contractile phenotypes to transcriptomic signatures
      This multi-parameter analysis enables comprehensive characterization of variant-specific contractile effects beyond simple binary classification.

How do experimental findings translate to clinical variant interpretation?

Experimental data provides crucial evidence for clinical variant classification:

  • Classification framework:

    • Significant deviation from wild-type in functional assays supports pathogenicity

    • Direction of change (increased vs. decreased function) predicts phenotype (HCM vs. DCM)

    • Magnitude of change may correlate with severity or penetrance

    • Concordance across multiple assays strengthens classification confidence

  • Experimental criteria for reclassification:

    • Variants previously classified as pathogenic/likely pathogenic should demonstrate significant functional effects

    • Variants of uncertain significance showing clear functional effects may be reclassified

    • Benign-like functional outcomes provide evidence against pathogenicity

  • Limitations and considerations:

    • Experimental evidence is one component of the ACMG classification framework

    • Some variants may affect protein functions not captured in current assays

    • The SarcTg platform cannot detect aberrant RNA-splicing phenotypes
      Research indicates that approximately 6.7% of previously classified pathogenic/likely pathogenic TNNT2 variants may be benign based on functional data, while 9.5% of variants of uncertain significance demonstrated pathogenic-like functional effects.

What are the primary challenges in functionally characterizing the complete catalog of TNNT2 variants?

Comprehensive functional characterization of all 384 TNNT2 variants in ClinVar faces several challenges:

  • Technical challenges:

    • Scalability of experimental approaches for hundreds of variants

    • Standardization of functional assays across laboratories

    • Integration of multiple functional readouts into meaningful classifications

  • Biological considerations:

    • Accounting for genetic background effects and modifiers

    • Addressing sex-specific and ethnicity-dependent variant effects

    • Capturing developmental and temporal aspects of variant pathogenicity

  • Interpretation challenges:

    • Establishing thresholds for pathogenicity in continuous functional readouts

    • Resolving discrepancies between different functional assays

    • Translating in vitro findings to clinical outcomes

  • Implementation strategies:

    • Prioritize variants based on population frequency and in silico predictions

    • Develop higher-throughput screening methods for initial classification

    • Perform detailed functional characterization on variants with ambiguous initial results
      Addressing these challenges will require collaborative efforts across research groups and standardized reporting of functional outcomes.

How might new technological approaches advance TNNT2 variant characterization beyond current methods?

Emerging technologies offer opportunities to enhance TNNT2 variant characterization:

  • Advanced genome editing approaches:

    • Base editing and prime editing for precise variant introduction without DNA breaks

    • Multiplexed CRISPR screens for simultaneous assessment of multiple variants

    • Inducible variant expression systems to study temporal effects

  • Enhanced cellular models:

    • Engineered heart tissues with defined cell composition and architecture

    • Advanced maturation protocols incorporating mechanical and electrical stimulation

    • Multi-cellular models including fibroblasts and endothelial cells to study tissue-level effects

  • Novel functional readouts:

    • Label-free contractility measurements using video microscopy and machine learning

    • Multi-parameter phenotyping with high-content imaging

    • Single-cell transcriptomics to capture cellular heterogeneity in response to variants

    • Proteomic approaches to study post-translational modifications and protein interactions
      These technologies will enable more comprehensive and physiologically relevant characterization of TNNT2 variants.

What approaches can address sex and ethnicity dependence in TNNT2 variant effects?

To address potential sex and ethnicity dependence of TNNT2 variant effects, researchers should:

  • Cell line diversity:

    • Develop the SarcTg platform in female or non-European ancestry cell lines

    • Create a panel of hiPSC lines representing diverse ethnic backgrounds

    • Generate paired male/female isogenic lines to study sex-specific effects

  • Experimental design considerations:

    • Include sex and ethnicity as variables in experimental analyses

    • Test established pathogenic variants across different genetic backgrounds

    • Specifically investigate variants like I211T and N269K reported predominantly in non-European populations

  • Data integration approaches:

    • Correlate in vitro findings with clinical data stratified by sex and ethnicity

    • Develop computational models accounting for genetic background effects

    • Establish collaborative networks to share data across diverse populations
      These approaches will help determine whether genetic background influences the functional consequences of TNNT2 variants and improve clinical interpretation across diverse populations.

How might TNNT2 functional genomics platforms inform therapeutic development?

The mechanistic insights gained from TNNT2 functional genomics can guide therapeutic strategies:

  • Targeted approaches based on functional mechanisms:

    • For HCM variants with increased calcium sensitivity: calcium-desensitizing agents

    • For DCM variants with decreased contractility: positive inotropic compounds

    • For variants affecting specific protein interactions: peptide or small molecule modulators

  • Screening platforms for therapeutic discovery:

    • Use the NPPB→tdTomato reporter to screen compound libraries

    • Identify molecules that normalize reporter activity for specific variants

    • Test candidates in cardiac microtissue contractility assays for functional validation

  • Precision medicine applications:

    • Develop variant-specific treatment protocols based on functional characterization

    • Predict individual responses to standard heart failure medications

    • Guide timing of interventions based on molecular mechanisms The sarcomere functional genomics platform provides both a deeper understanding of disease mechanisms and a screening system for therapeutic development, potentially enabling more personalized approaches to cardiomyopathy treatment.

Product Science Overview

Structure and Function

Cardiac Troponin T (cTnT) is a protein encoded by the TNNT2 gene in humans. It is a single, non-glycosylated polypeptide consisting of 305 amino acids with a molecular weight of approximately 36.4 kDa . The recombinant form of cTnT is produced in Escherichia coli and is often fused to a 20-amino acid His-tag at the N-terminus to facilitate purification .

The primary function of cTnT is to bind tropomyosin and anchor the troponin complex to the thin filament of the muscle fiber. This anchoring is essential for the regulation of muscle contraction in response to changes in intracellular calcium ion concentrations. When calcium ions bind to Troponin C (TnC), it induces conformational changes in the troponin complex, allowing the interaction between actin and myosin, leading to muscle contraction .

Clinical Significance

Mutations in the TNNT2 gene are associated with various cardiomyopathies, including familial hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) . These mutations can lead to altered troponin function, resulting in impaired muscle contraction and heart function. Measurement of cTnT levels in the blood is a widely used diagnostic marker for myocardial infarction (heart attack) and other cardiac injuries .

Applications

Recombinant human cTnT is used in various research and clinical applications, including:

  • Diagnostic Assays: Measurement of cTnT levels in blood samples to diagnose myocardial infarction and other cardiac conditions .
  • Research: Studying the molecular mechanisms of muscle contraction and the effects of mutations in the TNNT2 gene .
  • Drug Development: Screening potential therapeutic compounds that target the troponin complex and modulate muscle contraction .
Storage and Handling

Recombinant human cTnT is typically supplied as a sterile-filtered, colorless solution in a buffer containing 20 mM Tris (pH 8.0) and 10% glycerol . It should be stored at 4°C if used within 2-4 weeks or at -20°C for longer periods. For long-term storage, the addition of 0.1% human serum albumin or bovine serum albumin is recommended 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.