NAA50 exhibits broad substrate specificity, targeting tetrapeptides with N-terminal methionine followed by hydrophobic residues. Key findings include:
Peptide Substrate | Observed Ion | Molecular Formula | Theoretical Mass (m/z) | Observed Mass (m/z) | Error (ppm) |
---|---|---|---|---|---|
LAAA + Naa50 | [M(Ac) + Na]⁺ | C₁₇H₃₀O₆N₄Na | 409.2063 | 409.2055 | 8 |
MEAA + Naa50 | [M(Ac) + K]⁺ | C₁₈H₃₀O₈N₄SK | 501.1421 | 501.1418 | 3 |
MKAA + Naa50 | [M(Ac) + K]⁺ | C₁₉H₃₅O₆N₅SK | 500.1945 | 500.1940 | 5 |
MSAA + Naa50 | [M(Ac) + K]⁺ | C₁₆H₂₈N₄O₇SK | 459.1316 | 459.1310 | 6 |
Table 1: Mass spectrometric analysis of NAA50-catalyzed acetylation of synthetic peptides .
NAA50’s activity is modulated by auxiliary subunits:
NAA15 enhances NAA50 activity but is inhibited by HYPK through steric hindrance .
Autoacetylation: NAA50 exhibits lysine-ε-acetyltransferase activity, suggesting dual catalytic roles .
Pan-cancer analyses reveal NAA50 is overexpressed in most malignancies and correlates with poor prognosis:
NAA50 exhibits serotonin N-acetyltransferase (SNAT) activity, contributing to melatonin production:
Parameter | Value | Source |
---|---|---|
Kₘ (Serotonin) | 986 μM | |
Vₘₐₓ (Serotonin) | 1800 pmol/min/mg protein | |
Kₘ (N-alpha-acetyltransferase) | 37°C optimum temperature |
Table 2: Enzymatic parameters of NAA50’s dual acetyltransferase activity .
In vivo: Overexpression in rice enhances melatonin levels and osmotic stress tolerance .
Mechanism: Acetylates serotonin to N-acetylserotonin, a precursor for melatonin synthesis .
NAA50 is ubiquitously expressed, with cytoplasmic and nuclear localization observed in cancer cells . Tissue-specific expression includes:
Table 3: Tissue expression profile of NAA50 .
Human N-alpha-acetyltransferase 50 (NAA50) is a 169 amino acid cytoplasmic protein belonging to the acetyltransferase family and GNAT subfamily . It functions as a catalytic component of the ARD1A-NARG1 complex, displaying alpha acetyltransferase activity . NAA50 is encoded by a gene mapping to human chromosome 3q13.2 .
Methodologically, NAA50 can be studied as part of the NatE complex, which consists of Naa10, Naa50, and the auxiliary subunit Naa15 . It plays a critical role in co-translational modification of newly synthesized proteins in eukaryotic cells . Specifically, human Naa50 acetylates methionine followed by any residue except for proline, indicating its broad substrate specificity .
NAA50 protein is primarily localized to the nucleoli in human cells, as demonstrated through immunofluorescence studies . Research using the Human Protein Atlas database has shown that NAA50 protein can be visualized in the nucleoli of A-431 and U-2 OS cells using specific antibody staining techniques .
For visualization purposes, DAPI staining is used for nuclear localization (shown in blue), while NAA50 protein staining appears in green using specific antibodies . Interestingly, while subcellular localization studies show nucleolar concentration, immunohistochemistry results across cancer tissues demonstrate moderate to strong cytoplasmic staining, suggesting potential context-dependent localization patterns .
For experimental studies requiring purified NAA50, researchers can express recombinant human NAA50 protein with an N-terminal His-tag in E. coli expression systems . Purification is typically accomplished using conventional chromatography techniques, with product validation performed via SDS-PAGE to confirm protein identity and purity .
The recombinant protein can be used for various applications including enzymatic assays, crystallization studies, and protein-protein interaction analyses. When designing expression constructs, researchers should consider that human NAA50 consists of 169 amino acids, and the addition of affinity tags may affect certain functional properties .
Pan-cancer analysis reveals differential NAA50 expression patterns across cancer types, with significant overexpression in multiple cancers compared to normal tissues. Studies using the TIMER2.0 database demonstrated increased NAA50 expression in BLCA, CESC, COAD, ESCA, HNSC, LIHC, LUAD, LUSC, and STAD .
Additional analysis through SangerBox3.0 database of TCGA and GTEx datasets confirmed and expanded these findings to include GBM, GBMLGG, LGG, UCEC, BRCA, and numerous other cancer types . Interestingly, NAA50 was found to be downregulated in kidney-related cancers including KICH, KIRC, KIRP, and PCPG, suggesting tissue-specific regulatory mechanisms .
For comprehensive pan-cancer analysis, researchers should employ multiple database tools including TIMER2.0, SangerBox3.0, and GEPIA, along with validation through protein-level analysis using immunohistochemistry via resources like the Human Protein Atlas .
NAA50 significantly impacts cancer cell proliferation, particularly in lung adenocarcinoma. In vitro experiments using knockout models demonstrate that silencing NAA50 significantly inhibits proliferation rates in H1299 and PC9 lung adenocarcinoma cell lines .
For experimental validation, researchers can employ multiple complementary approaches:
Western blot analysis to confirm NAA50 knockdown efficiency
CCK-8 assay to evaluate cell proliferation rates over time
EdU assay to directly visualize and quantify cellular proliferation
These combined approaches have demonstrated that NAA50 downregulation significantly inhibits proliferation in lung adenocarcinoma cells, confirming computational predictions that NAA50 is involved in cell cycle regulation and proliferation .
NAA50 expression shows significant correlations with immune cell infiltration patterns across cancer types. Analysis using the TIMER2.0 database reveals that NAA50 expression is:
Positively correlated with bone myeloid-derived suppressor cell (MDSC) infiltration in multiple cancer types
Negatively correlated with natural killer T (NKT) cell infiltration across various cancers
Associated with altered infiltration levels of B cells, CD4+ T cells, CD8+ T cells, macrophages, and neutrophils specifically in LUAD
Additionally, correlation analysis between NAA50 expression and major histocompatibility complexes (MHCs) and chemokines using the TISIDB database provides further insights into NAA50's role in tumor immune microenvironment regulation .
These findings suggest NAA50 may influence tumor immune evasion mechanisms, making it a potential target for immunotherapy approaches.
Single-cell sequencing provides powerful insights into NAA50's cancer-related functions. Analysis using CancerSEA's single-cell sequence data reveals that NAA50 positively correlates with cell cycle and invasion processes in most tumors, with particularly prominent effects in LUAD .
Methodologically, researchers can leverage single-cell data to:
Identify co-expressed genes (top 50 positively and negatively correlated genes)
Perform GO analysis to identify biological functions (revealing NAA50's involvement in chromosome segregation, DNA replication, cell cycle phase transition, etc.)
Conduct KEGG pathway analysis showing enrichment in cell cycle, RNA transport, DNA replication, Fanconi anemia pathway, p53 signaling, and other critical pathways
This multi-layered analysis approach helps elucidate NAA50's mechanistic role in cancer progression, particularly its function in cell cycle regulation.
DNA methylation analysis of NAA50 provides important insights into its regulation in cancer. Research shows that the methylation level of NAA50 promoter is higher in ESCA, KIRC, LIHC, LUSC, PAAD, and SARC compared to adjacent normal tissues, while decreased in BRCA and HNSC .
This cancer-specific methylation pattern may explain differential expression levels and contribute to NAA50's role in tumorigenesis. DNA methylation involves the production of 5-methylcytosine by adding a methyl group to cytosine under DNMT catalysis .
For comprehensive understanding, researchers should consider that DNA methylation not only serves as an important marker for drug efficacy, survival rates, and cancer recurrence, but is also closely related to immune responses in various cancers . Integrating methylation analysis with expression and functional data provides a more complete picture of NAA50's role in cancer biology.
When designing experiments to study NAA50 knockdown effects, researchers should consider multiple technical approaches:
siRNA or shRNA approaches: For transient or stable knockdown of NAA50 in cell lines
CRISPR-Cas9 system: For complete knockout studies or precise genetic modifications
Validation of knockdown efficiency: Western blot analysis is essential to confirm reduced NAA50 protein levels
Selection of appropriate cell lines: Based on search results, H1299 and PC9 lung adenocarcinoma cell lines have been successfully used to study NAA50 function
Multiple functional assays: Employ CCK-8 for proliferation rate assessment and EdU assay for direct visualization of cellular proliferation
This comprehensive experimental design allows for robust assessment of NAA50's functional roles in cancer biological processes.
To investigate NAA50's role in cell cycle regulation, researchers should employ a multi-faceted approach:
Bioinformatic analysis: Examine correlations between NAA50 expression and CDK family, CDKI family, and Cyclin family genes using databases like TIMER2.0 (e.g., correlation with CDK1)
GO and KEGG pathway analysis: Identify enrichment in cell cycle-related pathways and biological processes, including chromosome segregation, mitotic cell cycle phase transition, and cell cycle checkpoints
Flow cytometry: For cell cycle profile analysis following NAA50 manipulation
Western blot analysis: Evaluate expression changes in key cell cycle regulators (cyclins, CDKs) after NAA50 knockdown
Single-cell analysis: Correlate NAA50 expression with cell cycle states across different cancer types
This integrated approach provides comprehensive insights into NAA50's mechanistic role in cell cycle regulation across cancer contexts.
For comprehensive NAA50 research, investigators should utilize multiple specialized databases and computational tools:
NAA50 shows considerable potential as a cancer biomarker based on several consistent findings:
NAA50's characteristics suggest significant potential as a therapeutic target:
Functional importance: NAA50 knockout significantly inhibits cancer cell proliferation, particularly in LUAD models
Pathway involvement: NAA50 regulates critical pathways including cell cycle, DNA replication, and p53 signaling
Immune correlation: NAA50 expression correlates with immune cell infiltration patterns, suggesting potential combination with immunotherapy approaches
Cancer-specific overexpression: Elevated in most cancer types while showing lower expression in normal tissues, potentially offering a therapeutic window
Therapeutic strategies might include development of small molecule inhibitors targeting NAA50's acetyltransferase activity, siRNA-based approaches for expression suppression, or targeting NAA50-dependent pathways. Further research should focus on identifying cancer types most dependent on NAA50 function and potential resistance mechanisms.
Several critical questions remain unanswered regarding NAA50:
Substrate specificity mechanism: While we know NAA50 acetylates methionine followed by any residue except proline , the structural basis for this specificity remains to be fully characterized
Tissue-specific regulation: The mechanisms explaining why NAA50 is downregulated in kidney cancers while upregulated in most other cancer types need investigation
Interaction with immune system: How NAA50 influences immune cell infiltration and potentially contributes to immune evasion needs deeper exploration
Post-translational modifications: How NAA50 itself is regulated through potential phosphorylation, ubiquitination, or other modifications remains largely unknown
Therapeutic resistance: Potential mechanisms of resistance to NAA50-targeted therapies need to be anticipated and studied
Addressing these questions would significantly advance our understanding of NAA50 biology and its therapeutic potential.
Integration of multiple omics technologies offers comprehensive insights into NAA50 function:
Genomics: Further analysis of NAA50 genetic variations, copy number alterations, and promoter variants across populations and disease states
Transcriptomics: RNA-seq and single-cell transcriptomics to understand cell-type specific expression patterns and regulatory networks
Proteomics: Mass spectrometry-based approaches to identify the complete set of proteins acetylated by NAA50 and how this acetylome changes in disease states
Epigenomics: Deeper investigation of NAA50 promoter methylation patterns and their functional consequences in different cancer types
Metabolomics: Investigation of how NAA50 activity impacts cellular metabolism, particularly in cancer contexts This integrated approach would provide a systems-level understanding of NAA50's role in human biology and disease, potentially revealing novel therapeutic opportunities and biomarker applications.
N Alpha-Acetyltransferase 50 (NAA50), also known as the NatE catalytic subunit, is a protein-coding gene that plays a crucial role in the acetylation of proteins. This enzyme is part of the N-terminal acetyltransferase (NAT) family, which is responsible for the co-translational modification of proteins by acetylating the N-terminus of nascent polypeptide chains. NAA50 is particularly significant due to its broad substrate specificity and its involvement in various cellular processes.
The NAA50 gene is located on human chromosome 3q13.2 . It encodes a protein that is composed of 169 amino acids with a molecular weight of approximately 19 kDa. The protein is characterized by its ability to acetylate the N-terminus of proteins that retain their initiating methionine . This acetylation process is essential for the stability, localization, and function of many proteins.
NAA50 exhibits both N-alpha-acetyltransferase and N-epsilon-acetyltransferase activities . The enzyme acetylates the initiator methionine of most peptides, except those with a proline in the second position . Additionally, it can acetylate the side chain of specific lysines on proteins, although the relevance of this activity in vivo remains unclear .
The acetylation process mediated by NAA50 is crucial for several cellular functions, including the establishment of mitotic sister chromatid cohesion and the regulation of protein-protein interactions . NAA50 is also involved in the acetylation of histone H4, which plays a role in chromatin remodeling and gene expression .
NAA50 is predominantly localized in the cytosol and nucleus of cells . It is a part of the NatA complex, which is associated with ribosomes where the acetylation reaction occurs co-translationally as the peptide extrudes from the ribosome . This localization is essential for its function in modifying newly synthesized proteins.
Mutations or dysregulation of the NAA50 gene have been associated with several diseases, including Chops Syndrome and Ogden Syndrome . These conditions are characterized by developmental delays, intellectual disabilities, and other systemic abnormalities. Understanding the role of NAA50 in these diseases can provide insights into potential therapeutic targets.