TPM2 Human (Tropomyosin 2) refers to the gene-encoded protein β-tropomyosin, a critical component of striated and smooth muscle function. The TPM2 gene (OMIM: 190990) is located on chromosome 9p13 and produces a 284-amino-acid coiled-coil protein that stabilizes actin filaments and regulates muscle contraction . Its primary role involves modulating actomyosin interactions in skeletal and cardiac muscles, with isoform-specific functions influencing muscle fiber type and calcium sensitivity .
Structure
β-Tropomyosin forms homodimers or heterodimers with α-tropomyosin (TPM1), adopting a bent coiled-coil conformation to bind actin filaments . The protein spans ~32 kDa, with phosphorylation sites (e.g., Serine-283) influencing polymerization and stress response .
Isoforms
Alternative splicing generates multiple isoforms, though TPM2β (muscle-specific) is predominant. TPM2α, TPM2δ, and TPM2η are less common, with TPM2β expression inversely correlated with malignancy in breast cancer cell lines .
Muscle Contraction Regulation
β-Tropomyosin modulates calcium-dependent actomyosin interactions via the troponin-tropomyosin complex. In diastole, it inhibits myosin binding to actin; during systole, calcium-induced conformational shifts release this inhibition . Overexpression studies in transgenic mice revealed delayed relaxation and reduced ventricular function when β-tropomyosin exceeds 75% of total tropomyosin .
Atherosclerosis: Reduced TPM2 expression correlates with plaque formation, suggesting a potential diagnostic marker .
Cancer: TPM2 upregulation in colon cancer-associated stromal cells (TASCs) promotes tumor growth and metastasis .
Pathogenic Variants
Over 28 TPM2 mutations disrupt tropomyosin flexibility, calcium sensitivity, or actin binding. For example:
V129A, E139K, and A155T reduce actomyosin ATPase activity and impair zebrafish/mouse muscle development .
Y265C increases calcium sensitivity, causing hypercontractility .
Mouse Overexpression: β-Tropomyosin >75% of total tropomyosin causes neonatal lethality and cardiac dysfunction .
Zebrafish: Pathogenic variants (e.g., E139K) disrupt muscle morphogenesis, correlating with human disease severity .
Cancer Insights
In breast cancer, TPM2β expression inversely correlates with malignancy, while TPM1λ (a novel TPM1 isoform) promotes transformation . In colon cancer, TPM2 upregulation in TASCs enhances tumor growth and metastasis .
Partner | Function | Source |
---|---|---|
Actin | Stabilization of filaments | |
Troponin T/C/I | Calcium-dependent contraction control | |
Myosin | Modulates ATPase activity | |
Caldesmon (smooth muscle) | Regulates contraction |
TPM2 (Tropomyosin 2) is an actin-binding protein central to muscle contraction regulation. Its sequence consists of periodic repeats corresponding to seven actin-binding sites, which are further divided into two functionally distinct halves . TPM2 plays a crucial role in regulating the interaction between myosin and actin in striated muscle contractility, fundamentally controlling how muscle cells generate force and movement .
TPM2 can be measured through several complementary methodologies:
Protein expression assessment: Immunohistochemical staining and grading is commonly employed in tissue samples, as demonstrated in studies examining TPM2 expression in 155 breast cancer tissues .
mRNA quantification: RNA-seq data analysis from databases like TCGA (The Cancer Genome Atlas) allows researchers to compare TPM2 expression across different tissue types and disease states .
Targeted expression analysis: RT-qPCR using specific primers (forward: 5'-AAGGGGACAGAGGATGAG-3' and reverse: 5'-CTTTCTCAGCCTCCTCCA-3') enables quantitative assessment of TPM2 expression in experimental settings .
Epigenetic regulation: Analysis of promoter methylation levels using databases like UALCAN helps understand regulatory mechanisms affecting TPM2 expression .
The most extensively studied isoform in skeletal muscle research is Tpm2.2, which is specifically expressed in striated muscle tissue . Mutations in this particular isoform have been linked to various myopathies with distinct contractile phenotypes. The functional specificity of this isoform makes it particularly important when investigating muscle disorders and contractility mechanisms .
TPM2 has emerged as a potential diagnostic and therapeutic biomarker for atherosclerosis based on multiple lines of evidence:
Differential expression: Bioinformatic analysis of expression profiling datasets (GSE43292 and GSE57691) identified TPM2 as a significantly differentially expressed gene between normal and atherosclerotic samples .
Decreased expression: TPM2 is displayed at lower levels in atherosclerosis models and clinical samples compared to normal tissue .
Validation studies: The relationship between TPM2 and atherosclerosis has been verified using both animal models and neural network prediction modeling approaches .
The downregulation of TPM2 in atherosclerotic tissue suggests it may play a protective role in normal vascular function, with its loss potentially contributing to disease pathogenesis .
Myopathy-linked mutations in Tpm2.2 have been categorized into two distinct groups based on their functional effects:
Hypercontractile mutations:
Hypocontractile mutations:
These mutations have opposite effects on product release kinetics during the actin-myosin cross-bridge cycle, directly correlating with the clinical muscle disease phenotypes observed in patients .
TPM2 shows significant clinical relevance in breast cancer:
Expression patterns: TPM2 is consistently downregulated at both mRNA and protein levels in breast cancer compared to normal breast tissue .
Epigenetic regulation: The promoter region of TPM2 shows significantly higher methylation in breast cancer tissues, suggesting epigenetic silencing as a potential mechanism for reduced expression .
Clinicopathological correlations: Lower TPM2 expression is significantly associated with:
This data is summarized in the following table:
Clinical Parameter | Total Cases | Low TPM2 Expression | High TPM2 Expression | P Value |
---|---|---|---|---|
Lymph node status | ||||
Negative | 89 | 28 (31.5%) | 61 (68.5%) | <0.001 |
Positive | 66 | 47 (71.2%) | 19 (28.8%) | |
Histological grade | ||||
I+II | 93 | 36 (38.7%) | 57 (61.3%) | 0.0084 |
III | 62 | 38 (61.3%) | 24 (38.7%) |
These findings suggest TPM2 functions as a potential tumor suppressor in breast cancer, with its loss associated with more aggressive disease features .
Researchers employ several sophisticated methodologies to study TPM2 function in muscle:
FRET (Fluorescence Resonance Energy Transfer): Used to examine binding geometry and orientation of Tpm2.2 on actin, revealing how mutations affect molecular positioning .
Actin-binding assays: Assess how wild-type and mutant TPM2 variants interact with actin filaments, providing insights into binding affinity and dynamics .
Myosin ATPase activity assays: Measure how TPM2 variants influence myosin's enzymatic function, a critical determinant of muscle contractility .
Single ATP turnover kinetic experiments: Evaluate how mutations affect product release rates during the myosin ATPase cycle, directly linking molecular changes to contractile phenotypes .
These complementary approaches allow researchers to connect structural alterations in TPM2 to functional consequences at the molecular, cellular, and tissue levels.
To investigate TPM2's role in cancer-immune cell interactions, researchers employ in vitro co-culture systems:
Macrophage differentiation protocol:
Co-culture system:
This methodology enables assessment of how tumor-associated macrophages might influence TPM2 expression in cancer cells, potentially contributing to disease progression mechanisms.
Computational methods play crucial roles in TPM2 research:
Differential expression analysis: Using RNA-seq data from TCGA and other repositories to identify expression patterns across tissues and disease states .
Functional annotation: Gene Ontology (GO) analysis identifies biological processes, molecular functions, and cellular components associated with TPM2 .
Pathway enrichment: KEGG and Gene Set Enrichment Analysis (GSEA) reveal signaling networks involving TPM2 .
Survival analysis: Kaplan-Meier plots correlate TPM2 expression with clinical outcomes .
Diagnostic evaluation: ROC (Receiver Operating Characteristic) curve analysis assesses TPM2's potential as a diagnostic biomarker .
Predictive modeling: Neural networks can validate relationships between TPM2 expression and disease phenotypes .
These approaches help researchers contextualize experimental findings within broader biological systems and translate basic research into potential clinical applications.
The functional architecture of TPM2 is critical to understanding its role in pathology:
Domain structure: TPM2 contains seven actin-binding periods divided into functionally distinct N-terminal and C-terminal halves .
Structure-function relationships: Mutations in N-terminal halves of periods 1 and 5 (D20H, E181K) create hypercontractile phenotypes by altering Tpm2.2 positioning on actin and enhancing myosin function .
Contrasting effects: Mutations in C-terminal halves of periods 1 and 5 (E41K, N202K) produce opposite effects, inhibiting motor activity .
This domain-specific functionality raises important research questions:
How do mutations in other actin-binding periods affect function?
Do specific domains interact with different regulatory proteins?
Can domain-specific therapeutic targeting be achieved to address specific disease phenotypes?
Multiple regulatory mechanisms likely influence TPM2 expression in cancer:
Epigenetic regulation: Promoter methylation levels are significantly higher in breast cancer tissues compared to normal breast tissue, suggesting epigenetic silencing as a key mechanism .
Transcriptional control: Analysis of transcription factor binding sites could reveal proteins regulating TPM2 expression that may be dysregulated in cancer.
Post-transcriptional mechanisms: miRNAs or RNA-binding proteins might contribute to reduced TPM2 levels in tumor tissues.
Advanced research questions include:
Can DNA methyltransferase inhibitors restore TPM2 expression in cancer?
What transcription factors are critical for tissue-specific TPM2 expression?
How does the tumor microenvironment influence TPM2 regulatory mechanisms?
The relationship between TPM2 and immune response represents an emerging research area:
Co-culture experiments: Studies examining TPM2 expression changes during macrophage-cancer cell interactions suggest immune-mediated regulation .
Research questions: Several critical questions remain unexplored:
Does TPM2 expression in cancer cells affect macrophage polarization?
Can infiltrating immune cells influence TPM2 methylation patterns?
Does TPM2-mediated cytoskeletal organization affect antigen presentation or immune synapse formation?
Is TPM2 expression associated with immunotherapy response in clinical settings?
Investigating these interactions could reveal new therapeutic approaches targeting the TPM2-immune axis in disease.
Several cutting-edge technologies show promise for TPM2 research:
Single-cell analysis: Single-cell RNA-seq and proteomics can reveal cell-specific TPM2 expression patterns and heterogeneity within tissues .
Gene editing approaches: CRISPR-Cas9 technology allows precise modification of TPM2 to study specific mutations or regulatory elements.
Advanced structural biology: Cryo-electron microscopy could provide detailed visualization of how TPM2 mutations affect protein structure and actin interactions.
Organoid models: Patient-derived organoids offer physiologically relevant systems to study TPM2 in disease contexts.
These technologies may help bridge the gap between molecular mechanisms and clinical manifestations of TPM2-related disorders.
TPM2 shows significant potential for clinical applications:
Biomarker development: TPM2 expression or methylation patterns could serve as diagnostic, prognostic, or predictive biomarkers in atherosclerosis and cancer .
Therapeutic targeting: Understanding the specific effects of TPM2 mutations could lead to personalized treatments for myopathies based on contractile phenotypes .
Risk stratification: TPM2 expression patterns might help identify high-risk patients who would benefit from more aggressive intervention or monitoring.
A multi-omics approach integrating genomic, transcriptomic, proteomic, and epigenomic data will likely provide the most comprehensive framework for translating TPM2 research into clinical practice.
Several obstacles remain in translating TPM2 discoveries to clinical applications:
Tissue-specific effects: TPM2 functions differently across tissue types, requiring context-specific research approaches.
Technical standardization: Development of standardized assays for TPM2 detection in clinical samples is needed for biomarker implementation.
Model systems: Creating physiologically relevant models that accurately recapitulate human disease features remains challenging.
Therapeutic specificity: Developing interventions that target disease-specific TPM2 functions without disrupting normal physiological roles requires sophisticated drug design.
Addressing these challenges will require multidisciplinary collaboration between basic scientists, clinicians, and biotechnology researchers to advance TPM2 from bench to bedside.
Tropomyosin-2, also known as TPM2, is a protein that plays a crucial role in muscle contraction and cytoskeletal functions. It is a member of the actin filament binding protein family and is predominantly expressed in slow, type 1 muscle fibers. The recombinant form of this protein, known as Tropomyosin-2 (Human Recombinant), is produced using recombinant DNA technology, which allows for the expression of the protein in a host organism such as Escherichia coli.
Tropomyosin-2 is composed of 284 amino acids and has a molecular weight of approximately 35.1 kDa. The protein is characterized by its ability to bind along the length of actin filaments, stabilizing them and regulating their interactions with other proteins. This binding is essential for the proper functioning of muscle contraction and other cellular processes that involve the cytoskeleton.
The recombinant form of Tropomyosin-2 is typically produced in Escherichia coli. The gene encoding the protein is inserted into a plasmid vector, which is then introduced into the bacterial cells. The bacteria are cultured under conditions that promote the expression of the recombinant protein. Once expressed, the protein is purified using conventional chromatography techniques to achieve a purity of over 90% .
Recombinant Tropomyosin-2 is used in various research applications, including studies on muscle physiology, cytoskeletal dynamics, and protein-protein interactions. It is also used in the investigation of diseases associated with mutations in the TPM2 gene, such as cap disease, nemaline myopathy, and distal arthrogryposis syndromes .
Mutations in the TPM2 gene can lead to a range of muscle disorders. These mutations can alter the expression and function of Tropomyosin-2, resulting in conditions such as cap disease, nemaline myopathy, and distal arthrogryposis syndromes. Understanding the structure and function of Tropomyosin-2 is therefore critical for developing therapeutic strategies for these diseases .