TNNI2 Human, His is the inhibitory subunit of the troponin complex, which regulates striated muscle contraction via calcium signaling:
Calcium Sensitivity: Binds calcium via troponin C (TnC), triggering conformational changes that expose myosin-binding sites on actin .
Actin-Tropomyosin Interaction: The C-terminal actin-binding domain (residues 156–178) inhibits actomyosin ATPase activity at low calcium levels .
Isoform Specificity: Exclusively expressed in fast-twitch skeletal muscles but also detected in smooth muscle and non-muscle tissues (e.g., corneal epithelium, cartilage) .
TNNI2 Human, His is produced via recombinant expression in E. coli and purified using nickel-chelating affinity chromatography:
Step | Details |
---|---|
Expression Host | E. coli |
Purification Method | Nickel-affinity chromatography (His-tag binding) |
Yield | ~0.25 mg/mL (concentration) |
Storage | Stable at -20°C; avoid freeze-thaw cycles |
TNNI2 mutations are linked to distal arthrogryposis type 2B (DA2B) and Sheldon-Hall syndrome, characterized by congenital joint contractures . Key mutations include:
Mutation | Effect | Source |
---|---|---|
p.Arg162Lys | Disrupts actin binding; causes variable DA2B phenotypes | |
p.R174Q, p.R156X | Impairs calcium-dependent inhibition; linked to arthrogryposis |
TNNI2 Human, His inhibits angiogenesis and tumor growth in ovarian carcinoma, acting as a coactivator of estrogen receptor-related receptor alpha (ERRα) .
Skeletal muscle injury releases fast TnI (fsTnI) into circulation, making it a potential biomarker for muscle damage .
Condition | Mechanism | References |
---|---|---|
DA2B | Missense/nonsense mutations in actin-binding domain → impaired inhibition | |
Sheldon-Hall Syndrome | TNNI2 mutations → hypercontractility and joint deformities |
TNNI2 overexpression inhibits angiogenesis in ovarian carcinoma, suggesting therapeutic potential .
Phosphorylation: Ser118 (AMPK substrate) modulates energy-dependent muscle function .
S-Glutathionylation: Cys133 modification enhances calcium sensitivity post-exercise .
Property | Value |
---|---|
Gene | TNNI2 (11p15.5) |
Protein Size | 202 aa (with His-tag) |
Isoelectric Point (pI) | 8.74 |
Stability | -20°C for long-term storage; 4°C for short-term use |
TNNI2 (troponin I2, fast skeletal type) is a gene encoding the fast skeletal muscle isoform of troponin I (fsTnI), which functions as the inhibitory subunit of the troponin complex in fast twitch skeletal muscle fibers . This 21.3 kDa protein consists of 182 amino acids including the first methionine and has an isoelectric point (pI) of 8.74 .
Methodologically, understanding TNNI2's role requires examining its function within the sarcomere, where it participates in the calcium regulation of muscle contraction and relaxation . The troponin complex, which includes troponin I (TnI), troponin T (TnT), and troponin C (TnC), controls the interaction between thick and thin filaments. When calcium levels are low, the troponin complex blocks binding between these filaments, preventing contraction. Increased calcium causes structural changes in the complex, exposing binding sites and enabling muscle contraction .
TNNI2 is one of three TnI isoforms in humans, each encoded by different genes:
Protein Isoforms | Slow Skeletal Muscle TnI | Fast Skeletal Muscle TnI | Cardiac TnI |
---|---|---|---|
Gene | TNNI1 | TNNI2 | TNNI3 |
Chromosomal Location | 1q31.3 | 11p15.5 | 19q13.4 |
Number of Exons | 9 | 8 | 8 |
Number of Amino Acids | 187 | 182 | 210 |
Molecular Weight (kDa) | 21.7 | 21.3 | 24.0 |
Isoelectric Point | 9.59 | 8.74 | 9.87 |
Tissue Specificity | Slow-twitch fibers, Embryonic heart | Fast-twitch fibers | Adult heart |
Methodologically, researchers can distinguish these isoforms through electrophoretic mobility, immunological techniques using isoform-specific antibodies, and molecular techniques targeting the unique regions of each isoform. From an evolutionary perspective, sequence analysis and co-evolutionary studies suggest that TNNI2 (fsTnI) represents the original TnI gene lineage, with TNNI1 and TNNI3 evolving later through gene duplication events .
For recombinant His-tagged TNNI2 expression, researchers should consider:
Expression System Selection: While E. coli is commonly used for its simplicity and high yield, mammalian expression systems may provide better post-translational modifications. For TNNI2, BL21(DE3) E. coli strains typically offer good expression levels.
Vector Design: Include a 6×His-tag (preferably N-terminal to avoid interference with C-terminal protein interactions) and consider adding a protease cleavage site if tag removal is required post-purification.
Culture Conditions: Induce expression at OD600 of 0.6-0.8 with 0.5-1mM IPTG at 25°C for 4-6 hours to balance yield and solubility.
Purification Protocol:
Lyse cells in buffer containing 50mM Tris-HCl (pH 8.0), 300mM NaCl, 10mM imidazole, 1mM DTT, and protease inhibitors
Purify using Ni-NTA affinity chromatography with imidazole gradient elution (50-250mM)
Further purify via ion-exchange chromatography (due to TNNI2's pI of 8.74 )
Final polishing with size-exclusion chromatography
Quality Control: Verify purity by SDS-PAGE and identity by Western blot with anti-His and anti-TNNI2 antibodies. Confirm activity through functional assays measuring calcium-dependent interactions with other troponin complex components.
To study TNNI2 mutations like c.525G>T (p.K175N) associated with distal arthrogryposis , researchers should employ a multi-faceted approach:
Patient Sampling and Genetic Analysis:
In Vitro Functional Studies:
Generate site-directed mutants in expression vectors
Express wild-type and mutant proteins in suitable cellular systems
Compare protein stability, folding, and degradation rates
Assess calcium binding affinity using fluorescence-based assays
Structural Analysis:
Use circular dichroism to assess secondary structure changes
Consider X-ray crystallography or NMR for detailed structural impacts
Employ molecular dynamics simulations to predict conformational changes
Muscle Fiber Studies:
Reconstitute troponin complexes with wild-type or mutant TNNI2
Measure force generation in skinned muscle fibers
Assess calcium sensitivity of the contractile apparatus
Animal Models:
Generate knock-in mice expressing the mutation of interest
Characterize the phenotype, focusing on limb development and muscle contractility
Use CRISPR/Cas9 to create models in other organisms for comparative studies
This comprehensive approach has proven effective in characterizing mutations like the p.K175N variant found in the Chinese DA2B family .
TNNI2 mutations are primarily associated with Distal Arthrogryposis Type 2B (DA2B), also known as Sheldon-Hall syndrome. The spectrum includes:
Missense Mutations:
Mutation Hotspots:
Most mutations cluster in functional domains involved in actin-tropomyosin interactions or calcium-dependent regulatory regions
Genotype-Phenotype Correlations:
For research purposes, a systematic approach to mutation analysis should include:
Whole-exome or targeted sequencing of TNNI2 in patient cohorts
Functional classification based on predicted protein domain disruption
In silico analysis of mutation impact on protein structure and function
Establishment of mutation databases with clinical correlations
TNNI2 mutations associated with distal arthrogryposis likely disrupt the normal calcium-dependent regulation of muscle contraction through several mechanisms:
Altered Inhibitory Function:
Disrupted Protein-Protein Interactions:
Calcium Sensitivity Changes:
Many mutations increase calcium sensitivity of the contractile apparatus
This creates a lower threshold for contraction initiation and may impair complete relaxation
Developmental Impact:
Altered contractile properties during embryonic development affect limb positioning
Sustained inappropriate contractions lead to joint fixation and the characteristic contractures
Research methodologies to investigate these mechanisms include:
In vitro motility assays comparing wild-type and mutant protein effects on actin filament movement
Calcium titration experiments measuring force generation in reconstituted systems
Structural studies examining conformational changes in the troponin complex
Single-molecule techniques to measure binding kinetics and force generation
The evolutionary history of TNNI2 provides significant insights into its functional specialization:
Evolutionary Origin:
Gene Duplication Events:
Co-evolution with TnT:
Functional Implications:
TNNI2's evolutionary position as the original isoform explains its specialized role in fast-twitch muscle
The divergence of TNNI1 and TNNI3 coincides with the evolution of specialized muscle types
These evolutionary relationships inform research approaches for comparative functional studies
Research methodologies examining evolutionary aspects should include:
Comparative genomics across species with varying muscle specializations
Analysis of conserved regulatory elements controlling tissue-specific expression
Investigation of how evolutionary changes in protein sequence relate to functional adaptations in different muscle types
TNNI2 undergoes several post-translational modifications (PTMs) that fine-tune its regulatory functions:
Phosphorylation Sites:
Functional Effects of Phosphorylation:
Altered binding affinity to other troponin components
Modified interaction with tropomyosin and actin
Changes in calcium sensitivity of the contractile apparatus
Adjustments in contraction-relaxation kinetics
Other PTMs:
Acetylation may occur at specific lysine residues
Oxidative modifications can affect protein function during oxidative stress
Potential ubiquitination sites regulate protein turnover
Methodological Approaches for PTM Research:
Mass spectrometry-based proteomics to identify modification sites
Phospho-specific antibodies for detecting specific phosphorylation states
In vitro kinase assays to determine kinetics and stoichiometry
Site-directed mutagenesis to create phosphomimetic or phospho-resistant variants
Functional assays comparing modified and unmodified protein properties
Research Challenges:
Distinguishing basal vs. stimulated phosphorylation states
Determining the combinatorial effects of multiple modifications
Correlating in vitro findings with physiological relevance
Cryo-electron microscopy (cryo-EM) offers revolutionary approaches for understanding TNNI2's structural dynamics:
Advanced gene editing technologies offer powerful tools for TNNI2 research:
CRISPR/Cas9 System for TNNI2 Studies:
Knock-in Strategies: Create precise mutations mirroring human variants (e.g., c.525G>T)
Design: Use homology-directed repair with donor templates containing desired mutations
Validation: Sequence verification, protein expression confirmation, and functional testing
Knockout Approaches: Generate complete or conditional TNNI2 knockouts
Design considerations: Potential embryonic lethality may require tissue-specific or inducible systems
Alternative strategies: Generate hypomorphic alleles with partial function
Cell Models:
iPSC-based Approaches:
Edit TNNI2 in induced pluripotent stem cells
Differentiate into skeletal muscle cells for functional studies
Advantages: Human genetic background, ability to create isogenic controls
Myoblast Cell Lines:
Edit C2C12 or primary human myoblasts
Assess effects during differentiation into myotubes
Measure contractile properties using micropatterned substrates
Animal Models:
Mouse Models: Generate knock-in lines with specific TNNI2 mutations
Zebrafish: Rapid model generation with ability to observe muscle development in vivo
Drosophila: Useful for high-throughput screening of multiple mutations
Analytical Approaches:
Transcriptomics to identify compensatory mechanisms
Proteomics to assess changes in the muscle interactome
Physiological measurements of muscle function
Developmental tracking to correlate with arthrogryposis phenotypes
Limitations and Considerations:
Potential off-target effects require thorough validation
Species differences in muscle development and function
Need for appropriate controls (including restoration of wild-type sequence)
Troponin I Type 2 is a critical component of the troponin complex, which plays a pivotal role in the regulation of muscle contraction in skeletal and cardiac muscles. The recombinant form of this protein, tagged with a His (histidine) tag, is widely used in research and diagnostic applications due to its ease of purification and high specificity.
Troponin I is one of the three subunits of the troponin complex, the other two being Troponin T and Troponin C. Troponin I is the inhibitory subunit that binds to actin in thin myofilaments to hold the troponin-tropomyosin complex in place, thereby inhibiting muscle contraction. When calcium ions bind to Troponin C, a conformational change occurs, leading to the displacement of Troponin I and allowing muscle contraction to proceed.
The recombinant form of Troponin I Type 2 is produced using Escherichia coli (E. coli) expression systems. This method involves inserting the gene encoding Troponin I into E. coli, which then produces the protein. The His tag, typically consisting of six histidine residues, is added to the N-terminus of the protein. This tag facilitates the purification of the protein using immobilized metal ion affinity chromatography (IMAC), which exploits the affinity of histidine residues for metal ions like nickel or cobalt .
Recombinant Troponin I Type 2 with a His tag is used in various applications, including: