TPGS2 exhibits aberrant expression across 33 cancer types, with significant variations between tumor and normal tissues:
Pan-cancer analysis (TCGA/GTEx data):
Overexpressed in 22 cancers (e.g., BRCA, GBM, LUAD).
TPGS2 expression correlates with survival outcomes across multiple malignancies:
TPGS2 demonstrates strong associations with tumor immunity:
Immune infiltration: Correlates with CD8+ T-cell density in COAD (r = 0.38, p = 0.002) and macrophage infiltration in LIHC (r = 0.42, p < 0.001) .
Immunomodulators: Expression linked to PD-L1, CTLA-4, and TIM-3 levels in multiple cancers .
Therapeutic targets:
Cell cycle regulation: Single-cell RNA sequencing reveals TPGS2 involvement in G2/M phase transition .
Metastasis: Co-expressed with MMP9 (r = 0.51, p < 0.001) and VEGFA (r = 0.49, p < 0.001) in pan-cancer analysis .
TPGS2 is an element of the neuronal polyglutamylase complex that plays a crucial role in the post-translational modification of tubulin. Specifically, it facilitates the addition of glutamate residues to C-terminal tubulin tails . This protein functions within the complex machinery responsible for tubulin polyglutamylation, which is essential for microtubule dynamics and stability. The gene encoding TPGS2 produces multiple isoforms through alternative splicing, which may contribute to tissue-specific functions. While initially identified for its role in neuronal function, recent research has uncovered significant implications for TPGS2 in various cancer types, suggesting a broader functional significance beyond its canonical role in tubulin modification .
Analysis of data from the GTEx database reveals that TPGS2 expression varies considerably across normal human tissues. The protein shows highest expression in testicular germ cell tissues (TGCT), bladder urothelial carcinoma (BLCA), and ovarian tissues (OV), while demonstrating lowest expression in liver hepatocellular tissues (LIHC) . This tissue-specific expression pattern suggests that TPGS2 may have distinct physiological roles depending on the tissue context. Understanding these baseline expression patterns is critical for interpreting pathological changes in TPGS2 expression during disease states, particularly in cancer development and progression .
TPGS2 exhibits abnormal expression in multiple cancer types compared to matching normal tissues. Integrated analysis of data from TCGA and GTEx databases demonstrates that TPGS2 mRNA is significantly upregulated in 22 cancer types, including breast cancer (BRCA), cervical squamous cell carcinoma (CESC), cholangiocarcinoma (CHOL), colorectal adenocarcinoma (COAD, READ), diffuse large B-cell lymphoma (DLBC), esophageal carcinoma (ESCA), glioblastoma multiforme (GBM), head and neck squamous cell carcinoma (HNSC), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), and stomach adenocarcinoma (STAD) . Conversely, TPGS2 is downregulated in adrenocortical carcinoma (ACC), kidney renal clear cell carcinoma (KIRC), acute myeloid leukemia (LAML), prostate adenocarcinoma (PRAD), and testicular germ cell tumors (TGCT) .
Immunohistochemistry (IHC) provides a reliable method for validating TPGS2 protein expression in cancer tissues. Analysis from the Human Protein Atlas (HPA) database confirms differential staining patterns between normal and cancerous tissues. Specifically, TPGS2 shows weak staining in normal liver, lung, colon, and stomach tissues, but displays stronger staining in their corresponding cancer tissues (LIHC, LUSC, COAD, and STAD) . Conversely, normal prostate and testes tissues exhibit medium TPGS2 staining, while their tumor tissues show weaker staining . These IHC results validate transcriptomic findings and provide visual confirmation of TPGS2 dysregulation in cancer, suggesting its potential utility as a diagnostic biomarker.
Genetic alteration analysis using the cBioPortal database with TCGA PanCancer Atlas data reveals cancer-specific patterns of TPGS2 alterations. Pancreatic adenocarcinoma (PAAD) exhibits the highest alteration rate (approximately 6%), with "amplification" and "deep deletion" as the predominant alteration types . Esophageal adenocarcinoma (ESAD) shows approximately 4% alteration frequency, primarily involving "deep deletion" . In contrast, endometrial carcinoma (UCEC) and skin cutaneous melanoma (SKCM) primarily display "mutation" as the main genetic alteration . Copy number alterations (CNAs) significantly correlate with TPGS2 mRNA expression levels across cancers, suggesting that genetic changes may drive abnormal expression patterns .
Single-cell sequencing data from the Cancer Single-cell State Atlas (CancerSEA) provides insights into TPGS2's functional associations at the cellular level. Analysis demonstrates that TPGS2 expression correlates with several critical cellular functions including cell cycle progression, metastatic potential, invasion capacity, inflammatory responses, and DNA damage processing . T-distributed stochastic neighbor embedding (t-SNE) visualization of single-cell expression profiles further elucidates cell-specific patterns of TPGS2 expression within heterogeneous tumor populations. This methodology allows researchers to identify specific cellular subpopulations where TPGS2 may play crucial roles, potentially revealing new therapeutic vulnerabilities or biomarker applications .
The Tumor Immune Estimation Resource (TIMER) database analysis reveals significant correlations between TPGS2 expression and the infiltration of various immune cell populations across multiple cancer types . Spearman correlation analysis demonstrates associations between TPGS2 mRNA expression and 20 distinct immune cell subsets, indicating a potential role for TPGS2 in modulating the tumor immune microenvironment . This relationship is particularly noteworthy given recent findings on the importance of B cells in the tumor microenvironment and their association with improved clinical outcomes in certain cancers. TPGS2 may influence the formation or function of tertiary lymphoid structures (TLSs) in tumor tissues, which are known to impact patient prognosis .
For progression-free survival (PFS), high TPGS2 expression is a risk factor in ACC, head and neck squamous cell carcinoma (HNSC), MESO, and UCS, while showing better PFS in lung carcinoma (LUCA) and pheochromocytoma and paraganglioma (PCPG) . Disease-specific survival (DSS) analysis identifies TPGS2 as a risk factor for ACC, LGG, LIHC, MESO, pancreatic adenocarcinoma (PAAD), and SARC, but as a protective factor for OV . These findings highlight the context-dependent prognostic significance of TPGS2 across different cancer types.
Spearman correlation analysis demonstrates significant associations between TPGS2 expression and established immunotherapy response biomarkers across various cancer types . Specifically, TPGS2 correlates with tumor mutational burden (TMB) and microsatellite instability (MSI), which are validated predictors of immunotherapy efficacy . Additionally, TPGS2 expression shows significant correlations with various immune regulators, including immunoinhibitors, immunostimulators, major histocompatibility complex (MHC) genes, chemokines, and chemokine receptors across different cancer types . These associations suggest that TPGS2 may serve as a complementary biomarker for predicting immunotherapy response, particularly in colon adenocarcinoma (COAD) and stomach adenocarcinoma (STAD), where the correlations are especially strong .
For robust analysis of TPGS2 expression data in cancer research, multiple statistical methodologies are recommended. The Wilcoxon rank-sum test is appropriate for evaluating statistical significance when comparing TPGS2 expression levels between tumor and normal tissues . For survival analyses, the Kaplan-Meier method with log-rank test provides visualization of survival differences, while univariate Cox regression quantifies hazard ratios associated with TPGS2 expression . When examining correlations between TPGS2 and other factors (immune cell infiltration, TMB, MSI), Spearman correlation analysis is the preferred approach . Statistical significance should be defined with standard thresholds (P<0.05, P<0.01, and P<0.001). Additionally, when analyzing transcriptomic data, the transcripts per million (TPM) format and log 2(TPM+1) transformation are recommended for expression profiling and subsequent analyses .
Several key databases and resources facilitate comprehensive investigation of TPGS2 in cancer research:
The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) via UCSC Xena (https://xenabrowser.net/) provide gene expression and clinical data for normal and cancer tissues .
The Cancer Cell Line Encyclopedia (CCLE) offers expression data across various cancer cell lines .
The Human Protein Atlas (HPA) (http://www.proteinatlas.org/) provides protein expression data and immunohistochemistry images for normal and cancer tissues .
The cBioPortal database (http://www.cbioportal.org) enables analysis of genetic alterations, including mutations, copy number variations, and their correlation with gene expression .
The Cancer Single-cell State Atlas (CancerSEA) provides single-cell sequencing data for functional state analysis .
The Tumor Immune Estimation Resource (TIMER) (http://timer.cistrome.org/) facilitates immune cell infiltration analysis .
Integration of data from these resources allows for multi-dimensional characterization of TPGS2's role in cancer biology.
Despite emerging evidence of TPGS2's importance in cancer, several knowledge gaps remain. The specific molecular mechanisms by which TPGS2 influences tumorigenesis and cancer progression are not fully elucidated . While correlations between TPGS2 and immune cell infiltration have been established, the causal relationships and underlying signaling pathways require further investigation . Additionally, the functional consequences of TPGS2 alterations on tubulin polyglutamylation in cancer cells and how these changes affect cellular processes like mitosis, migration, and response to therapy remain poorly understood. Future research should also address how TPGS2 interacts with other components of the tubulin modification machinery in the context of cancer .
To advance understanding of TPGS2 in cancer immunotherapy, several experimental approaches are recommended:
Functional studies using CRISPR-Cas9 gene editing to manipulate TPGS2 expression in cancer cell lines and assess effects on tumorigenicity, immune cell recruitment, and response to immunotherapeutic agents.
Development of patient-derived xenograft models with varying TPGS2 expression levels to evaluate immunotherapy response in vivo.
Spatial transcriptomics and multiplexed immunofluorescence imaging to characterize the relationship between TPGS2 expression and immune cell localization within the tumor microenvironment.
Mechanistic studies exploring how TPGS2-mediated tubulin modifications affect antigen presentation, immune checkpoint expression, and cytokine signaling.
Clinical correlation studies examining TPGS2 expression in pre- and post-treatment biopsies from patients receiving immune checkpoint inhibitors to validate its predictive value .
These approaches would significantly advance our understanding of TPGS2's role in cancer immunity and potentially lead to new therapeutic strategies.
Analysis using the tumor-immune system interaction database (TISDB) reveals significant associations between TPGS2 expression and molecular subtypes across various cancers . Notable correlations exist in breast cancer (BRCA), colorectal adenocarcinoma (COAD), head and neck squamous cell carcinoma (HNSC), brain lower grade glioma (LGG), lung squamous cell carcinoma (LUSC), and pheochromocytoma and paraganglioma (PCPG) . These associations suggest that TPGS2 may play distinct roles in different molecular subtypes of the same cancer type.
Furthermore, TPGS2 expression significantly correlates with immune subtypes in multiple cancers, including breast cancer (BRCA), kidney renal clear cell carcinoma (KIRC), liver hepatocellular carcinoma (LIHC), stomach adenocarcinoma (STAD), ovarian cancer (OV), and sarcoma (SARC) . These correlations highlight TPGS2's potential relevance in the context of tumor immunology and its potential utility as a biomarker for stratifying patients based on immune subtypes, which could inform immunotherapy treatment decisions .
Tubulin Polyglutamylase Complex Subunit 2 (TPGS2) is a crucial component of the neuronal polyglutamylase complex. This complex plays a significant role in the post-translational modification of tubulin, specifically through the addition of glutamate residues to the C-terminal tails of tubulin. This modification is essential for the proper functioning of microtubules, which are critical components of the cytoskeleton in eukaryotic cells.
The TPGS2 protein is involved in the polyglutamylation of tubulin, a process that affects the stability and function of microtubules. Polyglutamylation is a type of post-translational modification where glutamate residues are added to the gamma-carboxyl group of glutamate residues in proteins. This modification is particularly important in the regulation of microtubule-associated proteins and motor proteins, which are essential for intracellular transport and cell division.
The tubulin polyglutamylase complex, which includes TPGS2, is vital for the biogenesis and motility of cilia and flagella. These structures are essential for cell movement and sensory functions. In neurons, polyglutamylation of tubulin by TPGS2 is crucial for the proper functioning of axonal transport, which is necessary for the maintenance and function of neuronal cells .
The gene encoding TPGS2 is located on chromosome 15 in humans. It has been observed that there are multiple alternatively spliced transcript variants encoding different isoforms of this protein. This diversity in isoforms allows for the fine-tuning of tubulin polyglutamylation in various tissues and developmental stages .
Mutations or dysregulation of TPGS2 and the tubulin polyglutamylase complex can lead to various diseases. For instance, defects in this complex have been associated with ciliopathies, which are disorders caused by dysfunctional cilia. These conditions can lead to a wide range of symptoms, including respiratory problems, kidney disease, and retinal degeneration .
Recombinant TPGS2 is used in research to study the mechanisms of tubulin polyglutamylation and its effects on cellular functions. Understanding the role of TPGS2 in microtubule dynamics can provide insights into the development of therapeutic strategies for diseases related to ciliary dysfunction and neuronal transport .