Abstract The nucleolus is essential for ribosome biogenesis and stress regulation. However, because of its dynamic nature, there is still a lack of methods to specifically visualize nucleolar localization in living cells and to study dynamic changes in protein interaction networks within the cell nucleolus. In this study, we identified and engineered a signal peptide sequence, termed nucleolar beacon, which exhibits robust nucleolar localization and universal applicability across various mammalian cell types. Using this sequence, we established nucleolar indicator cell lines and demonstrated their practicality in studying nucleolar functions in living cells. In addition, by combining the signal peptide with proximity labeling technology, we developed an effective approach for capturing the nucleolar proteome and successfully identified nucleolar-associated proteins. These techniques provide effective and versatile tools for investigating nucleolar functions in living cells and offer a potential strategy for drug delivery applications. __________________________________________________________________ NoB serves as a reliable tool for visualizing nucleolar dynamics and identifying nucleolar-associated proteins. INTRODUCTION The nucleolus, the predominant membrane-less organelle in eukaryotic cells, plays a critical role in ribosome biogenesis and serves as a central hub for integrating internal and external signals associated with growth and stress ([42]1, [43]2). It exhibits a highly organized structure and liquid-like properties, which facilitate rapid adaptation to cellular homeostasis changes ([44]3). Damage to nucleoli, alterations in ribosomal proteins (RPs), and various stressors (such as cytotoxic agents, heat shock, nutrient deficiency, ultraviolet radiation, and viral infection) can lead to changes in nucleolar morphology ([45]4). These changes have been implicated in diverse diseases, including neurodegeneration (e.g., Parkinson’s disease), cancer (e.g., lung cancer and melanoma), and aging ([46]1, [47]5). Clinically, increased nucleolar size or number often signals poor prognosis in tumors due to heightened translational demands in rapidly dividing cells ([48]2, [49]6, [50]7). Understanding how external stimuli and intracellular factors influence the nucleolar dynamic alterations is thus critical for elucidating its role in disease. Traditional methods for studying nucleolar morphology and function, such as chemical staining, ribosomal RNA (rRNA) probes, and RP analysis, have yielded important insights but also have limitations ([51]8–[52]13). AgNOR staining, for instance, is commonly used to visualize the nucleolus and aid in the identification of tumor stage, grade, and aggressiveness ([53]10), yet it fails to fully capture nucleolar morphology. Recently developed rRNA-selective probes, including thioflavin T, 4MPS-TO, and low-energy red luminescent alkynylplatinum(II) complexes, offer rapid imaging of the nucleolus in live and fixed cells ([54]14–[55]16). Commercially available RNA stains such as Nucleolus Bright Red/Green (Dojindo), SYTO RNA Select (Thermo Fisher Scientific), and PhenoVue 512 (PerkinElmer) further expand this toolkit. However, these probes are constrained by nonspecific rRNA binding, poor water solubility, short emission wavelength, low stability against photobleaching, high cytotoxicity, elevated cost, spectral interference with other common fluorophores, and insufficient cellular membrane permeability. Fusion-based reporter systems such as green fluorescent protein (GFP)–RPL37 ([56]17), GFP-RPL29 and yellow fluorescent protein–RPS2 ([57]18), RPS9-HaloTag ([58]19), FBL-GFP ([59]20), upstream binding factor–HaloTag ([60]21), and enhanced GFP (EGFP)–DKC1 or eGFP–nucleophosmin 1 (NPM1) ([61]22) play a critical role in investigating the localization and activity of nuclear R proteins and are useful for identifying the location of the nucleolus. However, they did not fully address potential impacts on protein localization in other cellular compartments or the functional influence of the fusion tag protein. Mass spectrometry remains essential for nucleolar proteomics research but is hindered by the challenges of isolating nucleolar proteins. Currently, the isolation of nucleolar proteins is exclusively accomplished via density gradient centrifugation ([62]23, [63]24), which is labor intensive and prone to sample degradation. A recent innovation, light-induced targeting of endogenous condensates combined with the proximity labeling technique BiolD, has identified five notably enriched endogenous proteins associated with RNA polymerase II ([64]25). The nucleolus, as the most essential endogenous condensate in eukaryotic cells, is characterized by transient constituent components and poses significant challenges for investigation using conventional biological assays such as mass spectrometry and other proteomics profiling techniques. Therefore, it is imperative to develop straightforward, effective, and precisely focused approaches for monitoring the interaction network and dynamics of nucleolar proteins, particularly in the context of cell stress and cancer. In this study, we developed an effective approach for real-time nucleolar localization in live cells and pioneered a proximity labeling technique targeting the nucleolar proteome. By recombinantly modifying the putative nucleolar targeting sequence of Legionella pneumophila effector protein Ceg10, we successfully engineered a specific signal peptide [termed NoB (nucleolar beacon)] transported by importin-β (IMPβ) to the nucleolus. This peptide serves as a nucleolar marker for real-time cell research. In addition, we introduced a versatile proximity labeling system by fusing the NoB with biotin-catalyzing enzymes (UltraID and TurboID), which allowed the construction of a nucleolar protein interaction network. In conclusion, this study presents a user-friendly and intuitive technique to specifically identify the nucleolar localization in living cells while introducing an effective method to analyze the nucleolar protein interaction network and dynamic changes of nucleolar components in eukaryotic cells. It also proposes a potential strategy for the delivery of precision medicine for diseases associated with nucleolar dysfunction. RESULTS NoB: A robust nucleolus indicator for living cells The successful colonization of L. pneumophila depends on its ability to regulate host cell physiological functions through T4SS (type IV secretion system) effector proteins, some of which can penetrate the host nucleus ([65]26, [66]27). Previous studies have shown that Ceg10, a T4SS effector, inhibits host-conserved antimicrobial defenses ([67]28, [68]29). However, the precise physiological roles and host pathways regulated by Ceg10 remain unknown. In this study, subcellular localization experiments demonstrated that ectopically expressed EGFP-Ceg10 fusion proteins localized to the nuclei of human embryonic kidney (HEK) 293T cells (fig. S1A). The nuclear import of proteins generally involves nuclear localization signals (NLSs), linear motifs that bind specific karyopherin-β family of nuclear transport receptors to facilitate translocation. Sequence analysis using three independent NLS prediction tools confirmed that Ceg10 harbors NLS within its 26– to 64–amino acid region (fig. S1B). Notably, the amino acids 41 to 45 form a classical MP-NLS (monopartite NLS), characterized by clusters of positively charged residues such as arginine (R) and lysine (K) ([69]30, [70]31). Localization studies further revealed that the ectopically expressed truncated Ceg10[1–64] fused to the N terminus of EGFP was able to enter the nucleus and exhibited nucleolus localization. A similar pattern was observed with the truncated Ceg10[38–50] ([71]Fig. 1A). These findings indicate that the 38– to 50–amino acid region functions as a nucleolar localization signal (NoLS). To enhance nucleolar localization efficiency, we constructed tandem repeats of the NoLS peptide. All exogenously expressed EGFP fusion proteins were observed to be partially or fully localized in the same compartment as NCL (nucleolin), and this observation was supported by fluorescence colocalization analysis ([72]Fig. 1B). Moreover, we noted that the intensity of green fluorescence within the nucleolus increased proportionally with the number of concatenated sequences ranging from one to five repeats. However, no further intensity increase was observed beyond five repeats. Constructs fused to Flag–hemagglutinin (HA) or Myc tags retained nucleolar localization, confirming that the signal was functional regardless of tag type (fig. S1C). The 5× NoLS sequence, termed NoB ([73]Fig. 1C), consistently localized to the nucleolus in diverse mammalian cell lines, including A549, NIH3T3, PC9, and HeLa cells ([74]Fig. 1D). This establishes NoB as a robust and reliable indicator for nucleolar localization in mammalian cells. Fig. 1. Subcellular localization and nucleolar targeting of Ceg10 truncations and NoB in mammalian cells. [75]Fig. 1. [76]Open in a new tab (A) Representative fluorescence images of 293T cells transfected for 24 hours with EGFP, Ceg10[1–65]-EGFP, and Ceg10[38–50]-EGFP. (B) Subcellular localization and colocalization with NCL of one to six tandem recombinant NoLSs (Ceg10[38–50]) fused with EGFP at the C terminus after a 24-hour transfection in 293T cells. (C) Schematic illustration of the amino acid sequence and position of Ceg10[38–50] in the AlphaFold3-predicted structure of Ceg10, alongside the construction strategy for NoB. (D) Subcellular localization and colocalization of NoB-EGFP with NCL in NIH3T3, A549, PC9, and HeLa cells following a 24-hour transfection. Green fluorescence indicates the position of EGFP or EGFP-fused proteins, nuclear DNA was visualized with Hoechst staining (blue), and NCL was visualized using Cy3-conjugated anti-rabbit antibodies and NCL-specific rabbit antibodies (red). Yellow lines in the merged panels represent regions analyzed for spatial relationships between NoLS/NoB and NCL using ImageJ. Scale bars, 10 μm. KPNA1/KPNB1: Mediators of NoB nuclear translocation Classical NLSs typically bind IMPα proteins, which act as adaptors for IMPβ-mediated nuclear transport. To elucidate the nuclear import mechanism of NoB, we examined interactions between NoB and IMPα/β using coimmunoprecipitation (Co-IP) assays. The results demonstrated that NoB interacted with KPNA1, KPNA2, and KPNA6 of the IMPα family, as well as IMPβ/KPNB1 ([77]Fig. 2A). To further determine which nuclear transport proteins are critical for NoB transport, we reduced protein expression levels using small interfering RNA (siRNA) or short hairpin RNA (shRNA) and examined the distribution of NoB-EGFP through nuclear and cytoplasmic protein fractionation ([78]Fig. 2, B to E). The results showed that the knockdown of KPNA1 or KPNB1 significantly reduced the nuclear localization of EGFP-NoB, whereas the knockdown of KPNA2 or KPNA6 had minimal effect. Conversely, overexpression of KPNA1 and KPNB1 enhanced the nuclear localization of EGFP-NoB ([79]Fig. 2F). Immunofluorescence analysis following KPNA1 or KPNB1 knockdown further corroborated these findings ([80]Fig. 2, G and H). Collectively, these results indicate that the nuclear transport of NoB-EGFP primarily depends on KPNA1 and KPNB1, highlighting their roles as key mediators for nucleolar targeting. Fig. 2. Nuclear import of NoB is mediated by the IMPα/β. [81]Fig. 2. [82]Open in a new tab (A) Co-IP of NoB-EGFP and IMPα/β. (B to E) Protein expression levels of NoB-EGFP, KPNA1, KPNA2, KPNA6, and KPNB1 in nuclear and cytoplasmic fractions of 293T cells following siRNA- or shRNA-mediated knockdown. (F) Immunoblot (IB) analysis of NoB-EGFP, HA-KPNA1, and HA-KPNB1 expression in nuclear and cytoplasmic fractions after overexpression in 293T cells. LMNB2 (lamin-B2) and α-tubulin served as nuclear and cytoplasmic controls, respectively. (G and H) Immunofluorescence analysis of nucleolar localization and the fluorescence ratio of cytoplasm to nucleus analysis of NoB-EGFP following KPNA1 or KPNB1 knockdown in 293T cells for 24 hours. Scale bars, 10 μm. Green fluorescence indicates NoB-EGFP localization, nuclei were visualized with 4′,6-diamidino-2-phenylindole (DAPI) staining (blue), and Cy3-conjugated immunoglobulin G anti-rabbit antibodies were used to display the in situ KPNA1 and KPNB1 (red). Each dot represents one cell (n ≥ 90) from three independent experiments, and the data were presented as the means ± SD. siKPNA1 and siKPNA6, small interfering RNAs targeting KPNA1 and KPNA6, respectively. shKPNA2 and shKPNB1, short hairpin RNAs targeting KPNA2 and KPNB1, respectively. siControl, small interfering RNAs control. Data correspond to means ± 95% confidence and were analyzed using one-way analysis of variance (ANOVA), including all comparisons with Tukey’s correction. ***P < 0.001. 293T cell expressing NoB-EGFP and HeLa cell expressing NoB-EGFP: Reliable cell lines for investigation of nucleolar function Visualizing the localization, morphology, and dynamics of the nucleolus in living cells is crucial for studying the function of the nucleolus throughout the cell cycle. To accomplish this, we developed a lentivirus packaging expression plasmid to screen for cell lines consistently expressing the EGFP fusion NoB protein. Among the 30 strains tested, the most promising 293T and HeLa colonies, designated as TCN (293T cell expressing NoB-EGFP) and HCN (HeLa cell expressing NoB-EGFP), respectively, showed successful nucleolar localization. Immunofluorescence analysis revealed that NoB-EGFP localized to regions of the nucleolus marked by NPM1 and NCL, corresponding to the granular component (GC) and dense fibrillar component (DFC) regions ([83]Fig. 3A). In addition, Treacher Collins syndrome protein (TCOF1), a key regulator of RNA polymerase I that bridges ribosomal processing and modification enzymes, localizes to the fibrillar center (FC) region of the nucleolus ([84]32). The overlap of green fluorescence from NoB-EGFP with red fluorescence from TCOF1 further confirmed the FC localization of NoB-EGFP ([85]Fig. 3B). These observations indicate that NoB-EGFP is present in all three major nucleolar subregions. Fig. 3. Stable expression and nucleolar localization of NoB do not disrupt the physiological functions of TCN and HCN cells. [86]Fig. 3. [87]Open in a new tab (A and B) Fluorescence images showing NoB-EGFP localization in the nucleoli of TCN and HCN cells, colocalizing with NPM1 (red), NCL (purple), and TCOF1 (red), which mark GC, DFC, and FC of nucleoli, respectively. Green fluorescence indicates NoB-EGFP localization, nuclei were visualized with DAPI (blue), and NPM1, NCL, and TCOF1 were visualized using Cy3- or Cy5-conjugated secondary antibodies. Yellow lines in merged panels show regions analyzed for spatial relationships between NoB-EGFP and NPM1, NCL, or TCOF1 using ImageJ. Scale bars, 10 μm. (C) The ratio of the fluorescence of NoB in the nucleolus of TCN and HCN cells to that in the entire cell. Each dot represents the picture result of one field of different horizons (n = 20) from three independent experiments. (D) NoB-EGFP expression levels in TCN and HCN cells were determined by immunoblot assay, with overexpressed EGFP in 293T cells serving as the control. LMNB2 was used as an internal control. (E and F) Relative proliferation ratios over 24, 48, and 72 hours, determined via MTT assay. h, hours. (G and H) Representative flow cytometry results from the cell death assays. The ratio of total cell death was calculated by flow cytometry from three independent experiments. (I and J) Cell cycle overlay diagram of 293T or HeLa, 293T or HeLa supplemented with colchicine (0.125 μg/ml), and TCN or HCN cells were determined by flow cytometry. Percentages of cells in G[0]-G[1], G[2]-M, and S phases were analyzed by the ModFit software. Data are presented as means ± SD, with 95% confidence intervals. Statistical analyses were performed using one-way ANOVA with Tukey’s correction. P > 0.05 (n.s.), *P < 0.05, and ***P < 0.001. The statistical results of fluorescence localization analysis indicated that the proportion of fluorescence in the nucleolus could reach ~95% ([88]Fig. 3C). Western blot analysis showed that the expression levels of NoB-EGFP in TCN and HCN cells were lower than those of transiently expressed EGFP in 293T cells ([89]Fig. 3D). A 72-hour cell growth assay demonstrated no significant difference in proliferation ratio between TCN and 293T cells or HCN and HeLa cells ([90]Fig. 3, E and F). Flow cytometric analysis indicated that NoB-EGFP expression did not induce cell death ([91]Fig. 3, G and H). Furthermore, cell cycle analysis confirmed that TCN and HCN cells exhibited normal cell cycle progression compared with wild-type cells ([92]Fig. 3, I and J). Therefore, we successfully established the TCN and HCN cell lines, using NoB with nucleolar localization functionality as a reliable indicator of nucleolar morphology and integrity in living cells. Dynamic nucleolar changes observed in TCN and HCN cells To observe the alterations in the nucleolus indicated by NoB during mitosis in TCN and HCN cells, we monitored the fluorescence distribution every 6 hours after synchronizing the cells with colchicine. Different mitotic stages were successfully captured. The localization features of NoB-EGFP showed both similarities and differences between TCN and HCN at various stages of mitosis ([93]Fig. 4A). In TCN cells at the G[2]-S phase, NoB-EGFP fluorescence exhibited a rounder shape and a higher number of nucleoli compared to the G[0]-G[1] phase, whereas this difference was less pronounced in HCN cells. As mitosis began, during chromatin condensation, the green fluorescence of NoB-EGFP decreased. In TCN cells, NoB-EGFP was diffusely distributed throughout the cell, whereas in HCN cells, it was predominantly localized in the cytoplasm. Notably, NoB-EGFP did not exhibit the same chromosomal distribution pattern as NCL. Fig. 4. Dynamic changes of the nucleolus during mitosis and under stress conditions. [94]Fig. 4. [95]Open in a new tab (A) Fluorescence images of TCN and HCN cells at various mitotic stages after colchicine synchronization treatment. (B) Quantitative results concerning the number of nucleoli in TCN and HCN cells at different time points after synchronous treatment, based on a minimum of 40 cells per sample. (C) Micrographs of TCN and HCN cells after a 30-min infection with Lp02, A. baumannii, and S. aureus (multiplicity of infection = 20), followed by an additional 6-hour incubation period. (D to F) Quantification of nucleolus number, nucleolar area, and the nucleolar to nuclear area ratio in infected TCN and HCN cells (n ≥ 80). (G) Micrographs of TCN and HCN cells after a 5-hour treatment with actinomycin D (AMD), CX-5461, or CX-3543 (10 μg/ml). (H to J) Quantification of nucleolar number, nucleolar area, and the nucleolar to nuclear area ratio in drug-treated TCN and HCN cells (n ≥ 80). Green fluorescence indicates the position of NoB-EGFP, nuclei were visualized with DAPI staining (blue), NCL was visualized using Cy3-conjugated anti-rabbit antibodies and NCL-specific rabbit antibodies (red), and BF represents the bright-field image. Data are presented as the means ± SD, with confidence intervals of 95%. Statistical analyses were performed using one-way ANOVA with Tukey’s correction for multiple comparisons. Nonsignificant differences are denoted as n.s. (P > 0.05), while statistical significance is indicated as follows: *P < 0.05, **P < 0.01, and ***P < 0.001. h, hours. At metaphase, when chromosomes were preparing to divide, NoB-EGFP fluorescence nearly disappeared in both TCN and HCN cells until anaphase, when chromosomes were fully separated. At this stage, NCL fluorescence remained localized around the chromosomes. By the end of mitosis, as the cells formed two intact nuclei, NoB-EGFP reappeared within the nucleoli, colocalizing with NCL protein fluorescence. These observations were further supported by live-cell imaging of HCN cells using a confocal microscope, which confirmed the findings from the time-point analyses (movie S1). Quantitative analysis of nucleolar numbers at different time points revealed substantial fluctuations in TCN cells, ranging from one to nine nucleoli per cell ([96]Fig. 4B). The number of nucleoli increased 6 hours after synchronization, peaked at 12 hours, decreased to the initial level at 18 hours, and began to rise again at 24 hours. This fluctuation pattern suggests that a single mitotic cycle in TCN cells takes ~18 to 24 hours, with the S phase occurring around 12 hours postsynchronization. In contrast, HCN cells exhibited a more consistent range of nucleolar numbers (1 to 4), with slight increases observed at 6 and 18 hours. Furthermore, we investigated nucleolar changes in TCN and HCN following infections with L. pneumophila (strain Lp02), Acinetobacter baumannii, and Staphylococcus aureus. Both TCN and HCN cells exhibited an increased number of nucleoli after L. pneumophila or S. aureus infection ([97]Fig. 4, C to F). However, the nucleolar size in HCN cells was notably smaller compared to that in TCN cells. In contrast, A. baumannii infection led to an increase in nucleolar numbers in HCN cells but did not affect nucleolar numbers in TCN cells. Similar results were also observed in 293T and HeLa cells (fig. S2, A and B). In addition, we evaluated the effects of well-characterized nucleolar stressors, including actinomycin D (AMD), CX-5461, and CX-3543, on TCN and HCN cells ([98]Fig. 4, G to J). The results were consistent with previous findings, such as nucleolar disruption and alterations in nucleolar morphology ([99]33–[100]35). Similar results were also observed in 293T and HeLa cells (fig. S2, C and D). Collectively, these findings demonstrate that TCN and HCN are reliable cell lines for monitoring dynamic nucleolar changes, establishing NoB as an exceptional tool for nucleolar analysis. NoB-based proximity labeling: Approach for nucleolar proteomics in mammalian cells The conventional sucrose density gradient centrifugation method for exploring interactions between nucleolar proteins is hindered by specificity and labor-intensive protocols. Addressing this limitation, we developed an effective approach that combines the nucleolar localization capability of NoB with proximity labeling technology. We constructed a NoB-fused biotin-catalyzing enzyme expression vector and transfected it into 293T cells. Fluorescence analysis confirmed that both UltraID-NoB-EGFP and TurboID-NoB-EGFP localized to the nucleolus ([101]Fig. 5A). This localization pattern was consistent across other cell types, including HeLa, PC9, A549, and NIH3T3 cells (fig. S3). Fig. 5. Proximity labeling of the nucleolar proteome by NoB-mediated nuclear import of UltraID. [102]Fig. 5. [103]Open in a new tab (A) Fluorescence images of UltraID-NoB-EGFP localized to the nucleolus in 293T cells. Green fluorescence indicates the position of UltraID-EGFP or UltraID-NoB-EGFP, nuclei were visualized with Hoechst stain (blue), and NCL was visualized using Cy3-conjugated anti-rabbit antibodies and NCL-specific rabbit antibodies (red). (B) Experimental schematic of the procedure for UltraID-NoB-EGFP proximity labeling nucleolar proteome. (C) Immunoblot analysis of streptavidin affinity pull-down of biotinylated proteins from 293T cells expressing UltraID-EGFP or UltraID-NoB-EGFP. (D to F) Volcano plot, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway classification, and the top 20 Reactome enrichments of differentially notable proteins in the nucleolar proteome obtained through UltraID-NoB-EGFP proximity labeling. Sig., significance; GTP, guanosine 5′-triphosphate. (G to I) KPNA1 and KPNB1 mediate nuclear translocation of UltraID-NoB-EGFP. The protein expression levels of UltraID-NoB-EGFP, HA-KPNA1, and HA-KPNB1 in nuclear and cytoplasmic fractions of 293T cells were determined by immunoblot assay following their overexpression (G) or siRNA/shRNA-mediated knockdown (H and I). LMNB2 and α-tubulin served as nuclear and cytoplasmic controls, respectively. h, hours. Among the constructs, we selected UltraID-NoB-EGFP for its ability to catalyze rapid biotinylation ([104]36), and we used it to investigate the nucleolar proteome ([105]Fig. 5B). Western blot analysis demonstrated that UltraID-NoB-EGFP effectively biotinylated surrounding proteins, which could be enriched using streptavidin-coupled agarose beads ([106]Fig. 5C). Compared to the control UltraID-EGFP, the molecular weight distribution of biotinylated proteins was notably different. Mass spectrometry analysis identified 252 significantly enriched proteins (P < 0.05) (data S1), including 62 up-regulated [log[2] fold change (log[2]FC) > 1 and P < 0.05] ([107]Fig. 5D). Notably, many up-regulated proteins included ribosomal subunits such as PRL23, PRL4, PRL21, PRS16, PRS3A, PRL19, etc. Other enriched proteins included NOP56 and NOP58 (localized in the FC) and NCL (localized in the DFC) (data S1). Twenty up-regulated proteins were entirely absent in the control group, suggesting potential nucleolus-specific roles. Examples include ENO1, JPT2, RBM10, TTC1, and TMED10. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that these proteins were involved in diverse signaling pathways, with “translation” being the most abundant category under genetic information processing ([108]Fig. 5E). Reactome pathway analysis further highlighted significant enrichment in RNA metabolism, protein translation, and ribosomal small subunit pathways ([109]Fig. 5F). Mass spectrometry also identified nuclear transport proteins KPNA1, KPNA3, IPO9, CSE1L, and KPNB1. Co-IP experiments revealed that NoB predominantly interacts with KPNA1 and KPNB1, consistent with prior findings ([110]Fig. 2 and fig. S4A). Overexpression of KPNA3, IPO9, and CSE1L did not enhance the nuclear localization of NoB-EGFP in 293T cells (fig. S4B). To determine whether the nuclear transport of UltraID depends on NoB, we adopted the same methods of overexpression and knockdown of KPNA1 and KPNB1. Further validation showed that the nuclear localization of UltraID-NoB depended on KPNA1 and KPNB1. Knockdown of these proteins reduced the nuclear levels of UltraID-NoB, while their overexpression increased nuclear accumulation ([111]Fig. 5, G to I). In conclusion, we developed an effective method for capturing the nucleolar proteome of mammalian cells by combining NoB with proximity labeling. This approach relies on the nuclear import trafficking mediated by KPNA1 and KPNB1, offering an effective tool for nucleolar proteomics and expanding the understanding of nucleolar functions. DISCUSSION The nucleolus, a vital membrane-less structure within the eukaryotic nucleus, plays a crucial role in cell growth and the regulation of stress responses. However, because of its dynamic nature, there is still a lack of methods to efficiently visualize nucleolar localization in living cells and to study dynamic changes in protein interaction networks within the cell nucleolus. In this study, we explored the subcellular localization of the L. pneumophila effector Ceg10 and identified the short peptide Ceg10[38–50] capable of nucleolar localization as anticipated by NLS or NoLS prediction websites ([112]Fig. 1A and fig. S1, A and B). The central attribute of the sequence is the characteristic MP-NLS motif (RRSKKR), which is flanked by glutamate residues and features a lysine residue at the terminal end of the Ceg10[38–50] sequence, resulting in a peptide rich in polar amino acids. Upon cleavage of Ceg10[38–50] into two shorter peptides (QERRSKKR and ELHCK), there was a substantial reduction in nucleolar localization efficiency, especially for Ceg10[46–50], underscoring the necessity of the intact sequence for functionality (fig. S1D). After five consecutive repetitions of NoLS (NoB), the efficiency of nucleolar entry is maximized, allowing it to penetrate the nucleoli of various mammalian cells ([113]Fig. 1, B to D). The enrichment of lysine and arginine is a fundamental feature of NLSs. The P1 position of the classical MP-NLSs is invariably occupied by lysine [K(R/K)X(R/K)] ([114]37, [115]38). On the other hand, NoLSs are characterized by a higher proportion of these two amino acids and longer sequence length compared to NLSs ([116]39, [117]40). Numerous studies have demonstrated that NoLSs facilitate the accumulation of proteins in the nucleolus through a charge-dependent mechanism rather than a sequence-specific mechanism ([118]39, [119]41, [120]42). This view is supported by the analysis of the general nucleolar protein properties of Trypanosoma brucei, which identified basic amino acids as the key protein feature associated with nucleolar partition ([121]43). Recent research has revealed that nucleolar proteins contain D/E tracts, K blocks + E-rich regions, and proteins harboring high-scoring K blocks + E-rich regions are enriched in the FC/DFC, followed by GC ([122]44). The NoB displays the aforementioned characteristics, in addition to the presence of two E’s along with their relatively high content of the positively charged amino acids. The formation of K blocks and E-rich regions contributes to nucleolar localization. Furthermore, AlphaFold3 predicts that NoB cannot form a stable secondary structure. Previous studies have demonstrated that interactions mediated by disordered regions promote the specific localization of proteins to condensates ([123]38, [124]45). Therefore, the nucleolar localization of NoB is probably achieved through its high proportion of polar amino acids, K blocks, E-rich regions, and conformational flexibility. We further investigated the potential pathways for NoB nuclear entry. Classical NLSs typically contain four to eight positively charged residues that interact with the IMPα proteins, serving as adaptors for transport to the nucleus by IMPβ ([125]31, [126]38, [127]46, [128]47). In 293T cells, interaction studies between NoB and IMPα/IMPβ proteins revealed that NoB interacts with KPNA1, KPNA2, KPNA6, and KPNB1 ([129]Fig. 2A). However, overexpression and knockdown of these transporters showed that KPNA1 and KPNB1 had the most notable impact on the nuclear-cytoplasmic separation assay, while the effects of KPNA2 and KPNA6 were less pronounced ([130]Fig. 2, B to F). Immunofluorescence analysis further corroborated these findings, which indicate that the nuclear transport of NoB-EGFP depends on KPNA1 and KPNB1 ([131]Fig. 2, G and H). As the nuclear import–related proteins KPNA1, KPNA3, IPO9, and CSE1L were identified in the nucleolus mass spectrometry results, we also analyzed their interactions with NoB. These analyses indicated that NoB did not bind to KPNA3, IPO9, or CSE1L (fig. S4A). Compared to KPNA1 and KPNB1, overexpression of KPNA3, IPO9, and CSE1L did not significantly enhance the nuclear localization of NoB (fig. S4B). To eliminate the difference in nuclear transport mechanism caused by the difference in the length of the signal peptide and the protein carried by it, we monitored the nuclear-to-cytoplasmic protein ratio after knocking down or overexpressing KPNA1 and KPNB1 in cells overexpressing UltraID-NoB-EGFP. The results were consistent with those observed for NoB ([132]Fig. 5, G to I). Together, the translocation of NoLS into the nucleus is mainly mediated by KPNA1 and KPNB1. The Human Protein Atlas ([133]proteinatlas.org) shows that KPNA1 is detectable in all cell lines ([134]48). This characteristic is likely a key reason for NoB’s nucleolar localization across various mammalian cell types. Given the strong nucleolar localization properties of NoB, we established 293T and HeLa cell lines (referred to as TCN and HCN, respectively) stably expressing EGFP-NoB. These cell lines maintained normal physiological functions (e.g., growth, division, and apoptosis) and exhibited dynamic changes in nucleolar localization during the cell cycle ([135]Figs. 3 and [136]4, A and B, and movie S1). We observed that the number of nucleoli exhibited periodic fluctuations, with HCN cells showing smaller amplitude variations and shorter fluctuation periods compared to TCN cells. This characteristic aligns with the higher proliferative capacity of cancer cells. Exposure to well-characterized nucleolar stressors induced the expected nucleolar stress phenotypes ([137]Fig. 4, G to J), further validating the reliability of TCN and HCN cells for monitoring dynamic nucleolar changes. Another intriguing finding was the distinct changes in nucleolar number and morphology following infections with L. pneumophila, A. baumannii, and S. aureus ([138]Fig. 4, C to F). The observations underscore the critical role of NoB in detecting nucleolar stress responses induced by bacterial infections. These results also highlight the differences in stress responses between TCN and HCN cells to various pathogens, suggesting potential disparities in the regulation of nucleolar stress responses between normal and cancer cells. In future studies, we aim to further investigate the specific regulatory mechanisms underlying these phenomena. Compared to traditional nucleolar tracking methods, such as fluorescent protein–based reporters, rRNA probes, or nucleolar dyes, NoB exhibited high specificity and avoided nonspecific cytoplasmic targeting. Its versatility also enables researchers to establish additional nucleolar localization indicator cell lines. Therefore, these findings highlight the potential application of NoB as a precise indicator of nucleolar localization in living cells. Proteomic analysis of nucleolar proteins is essential for functional studies. In recent years, proximity labeling techniques have been widely applied across various aspects of proteomics research, facilitating the capture of transient or weak protein interactions ([139]36, [140]49–[141]51). In this study, we were surprised to find that NoB has the ability to transport various tags and proteins into the nucleolus, including biotin-catalyzed enzymes such as UltraID and TurboID ([142]Fig. 5A and fig. S3). By integrating NoB with UltraID, we successfully captured the nucleolar proteome, and the pathway enrichment analysis further validated its specificity to target nucleolar proteins ([143]Fig. 5, B to F). The notably different proteins encompassed a large number of ribosome proteins, such as PRL23, PRL4, PRL21, PRS16, PRS3A, PRL19, etc. In addition, key proteins localized to the FC region of the nucleolus, including NOP56 and NOP58, the DFC region, such as NCL, and the nucleolar periphery, such as DEAD-box helicase 21 (DDX21), were also identified. These findings demonstrate the effectiveness of this method for capturing the nucleolar proteome. We further evaluated the localization properties of UltraID-NoB-EGFP and TurboID-NoB-EGFP in other cell lines, confirming their versatility in mammalian cells (fig. S3). Collectively, this research highlights the potential of combining NoB with proximity labeling as a tool for analyzing the dynamic nucleolar proteome in mammalian cells. In this study, we identified and engineered a signal peptide sequence that exhibits nucleolar localization functionality and versatility in mammalian cells. Subsequently, we established nucleolar indicator cell lines for functional investigations in living cells. Furthermore, the combination of signal peptide and proximity labeling technology provided an effective approach to obtain the nucleolar proteome, successfully capturing the nucleolar-associated proteome ([144]Fig. 6). These techniques offer effective tools for investigating nucleolar functions in living cells and present a potential application method for drug delivery. Fig. 6. Schematic diagram of NoB as a tool for monitoring nucleolar morphology and proteomics. [145]Fig. 6. [146]Open in a new tab (Left) NoB-EGFP is transported into the nucleus through the combination of KPNA1 and KPNB1 and eventually reaches the nucleolus, where it can be used for nucleolar monitoring in living cells. (Right) The biotin catalytic enzyme UltraID bound to NoB-EGFP is transported into the nucleus via KPNA1 and KPNB1 and lastly reaches the nucleolus for the proximity labeling of the nucleolar proteome. In conclusion, we designed a signal peptide sequence, NoB, that exhibits strong nucleolar localization functionality and universal applicability in mammalian cells. Using NoB, we established nucleolar indicator cell lines for studying nucleolar functions in living cells. Furthermore, by combining NoB with proximity labeling technology, we successfully captured the nucleolar proteome ([147]Fig. 6). These techniques provide effective and versatile tools for investigating nucleolar functions in living cells and present a potential strategy for drug delivery applications. MATERIALS AND METHODS Cell culture HEK293T/293T, NIH3T3, HeLa, A549, and PC9 cell lines were cultured in Dulbecco’s modified Eagle’s medium (VivaCell, C3113-0500) supplemented with 10% fetal bovine serum (VivaCell, 04-001-1A), penicillin (50 U/ml), and streptomycin (50 mg/ml; VivaCell, 03-031-5B). Cells were maintained at 37°C in a humidified incubator with 5% CO[2]. Mycoplasma contamination was checked using a universal detection kit (Beyotime Biotechnology, C0297S). Plasmid constructions For ectopic expression in mammalian cells, full-length Ceg10, Ceg10[38–50], mutant variants, and tandem repeats (1 to 6× Ceg10[38–50]) were cloned into the pcDNA3.1 vector with N- or C-terminal EGFP, Myc, or Flag-HA tags. Biotinylation enzymes (UltraID and TurboID) fused with NoB-EGFP were also inserted into the pcDNA3.1 vector for expression in mammalian cells. For lentivirus packaging screening, NoB with an N-terminal EGFP tag was inserted into pCS-CG. The construction of these plasmids was accomplished using the homologous recombination method as per the ClonExpress Ultra One Step Cloning Kit protocol (Vazyme, C115-01). All plasmids and primers used are listed in tables S1 and S2. Transfection and Co-IP Plasmids were transfected into cells at 80% confluence using Lipo8000 (Beyotime, C0533) following the manufacturer’s protocol. After 24 hours, the cells were lysed in Western blot and IP cell lysis buffer (Beyotime, P0013) at 4°C for 20 min. Lysates were centrifuged at 2500 rpm for 10 min at 4°C, and the supernatant was incubated with Protein A + G Magnetic Beads (Beyotime, P2108) preconjugated with GFP-specific antibodies for 1 hour at room temperature. The beads were washed five times with precooled lysis buffer, and bound proteins were eluted, resolved via SDS–polyacrylamide gel electrophoresis (PAGE), and analyzed by immunoblotting using antibodies listed in table S3. Western blotting Cells were lysed at 4°C for 30 min with the radioimmunoprecipitation assay (RIPA) buffer (Beyotime, P0013B). Lysates were centrifuged at 12,000 rpm, and the supernatant was subjected to SDS-PAGE. Proteins were transferred to membranes and analyzed by immunoblotting using antibodies against GFP, HA, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), α-tubulin, lamin-B2, KPNA1, KPNA2, KPNA3, KPNA6, and KPNB1 (table S3). Immunofluorescence After 24 hours of plasmid transfection, cells were washed with phosphate-buffered saline (PBS) and fixed in 4% paraformaldehyde for 30 min at room temperature. Following three washes with PBS, the cells underwent permeabilization using PBS–Triton X-100 (0.5%, v/v) for 10 min. The samples were incubated overnight at 4°C with antibodies (1:200 dilution in 5% bovine serum albumin; refer to table S4 for antibody details) and subsequently with the fluorescence-labeled secondary antibodies for 60 min at room temperature. Last, staining of the cells was carried out using Hoechst 33342 (1:1000; Beyotime, C1025), followed by three washes with PBS and the addition of 200 μl of fluorescence decay-resistant sealing tablets. The fluorescence microscope (Leica, SP8) was used to analyze Hoechst and immunofluorescence signals, and the images were processed with the ImageJ software ([148]52). Stable cell line establishment The NoB-EGFP sequence was cloned into the pCS-CG vector and cotransfected with packaging plasmids psPAX2 and pMD2.G into 293T cells at a 4:3:1 ratio. After 48 hours, the supernatant was collected and filtered through a 0.45-μm filter membrane. Subsequently, 293T and HeLa cells were infected with 4 ml of the viral supernatant, followed by the addition of Polybrene (1 μg/ml) to enhance infection. After incubation for 6 hours, fresh medium was added, and cells were cultured for an additional 48 hours. Monoclonal cell lines exhibiting fluorescence were sorted using a flow cytometer. MTT assay The cytotoxicity of NoB-EGFP in TCN and HCN cell lines was evaluated using the MTT assay. The cells were trypsinized and seeded onto 96-well plates at a density of 5 × 10^3 cells per well. After an overnight incubation, the cells were designated as 0-hour samples. Subsequently, cells cultured for 24, 48, and 72 hours were collected, and 10 μl of MTT solution (5 mg/ml) was added to each well and incubated for 4 hours. After removing the medium, the formazan formed in the cells was dissolved using 150 μl of dimethyl sulfoxide by shaking at 60 rpm for 10 min at room temperature. The absorbance optical density values were then determined at a wavelength of 490 nm using a multimode reader (Molecular Devices, SpectraMax P1). The proliferation cell ratio was calculated using the following formula: optical density at 490 nm (OD[490]) of samples/OD[490] of wild-type HEK293T and HeLa at time point zero. Flow cytometric analysis of cell death and cell cycle Cell death and cell cycle detection were carried out following the manufacturer’s protocol of the Apoptosis and Necrosis Assay Kit (Beyotime, C1056) and Cell Cycle Kit (Beyotime, C1052). After incubation in six-well plates for 24 and 48 hours, cells were trypsinized and centrifuged at 1000g for 5 min at 4°C, followed by three washes with PBS. Subsequently, the cells were resuspended in 200 μl of 1× binding buffer containing 5 μl of Hoechst and 5 μl of propidium iodide (PI) and incubated for 20 min at room temperature. For cell cycle analysis, trypsinized cells were centrifuged at 1000g for 5 min at 4°C, washed three times with PBS, and fixed overnight at 4°C in 75% ethanol solution. The cells were then washed again thrice with PBS, gently resuspended in 195 μl of binding buffer, and stained with 5 μl of PI before being incubated at room temperature in the dark for 20 min. Last, the samples were analyzed using flow cytometry (Beckman Coulter, Gallios), and the data were processed using the Kaluza and ModFit analysis software to obtain accurate results. Gene knockdown To suppress the expression of KPNA1, KPNA2, KPNA3, KPNA6, KPNB1, and KPNB2, siRNAs targeting KPNA1 and KPNA6 were synthesized by General Biol (China). shRNAs were constructed in the pLV3 vector. HEK293T cells were cultured in six-well plates and cotransfected with NoB plasmids along with siRNA (100 nmol/ml) or PLV3-shRNA using Lipo8000 (Beyotime, C0533). Proteins were analyzed by SDS-PAGE and immunoblotting assays after 48 hours. The sequences for all siRNAs or shRNAs are listed in table S4. Subcellular fractionation The cytoplasmic and nuclear fractions were isolated using the protocol provided by the Nuclear and Cytoplasmic Protein Extraction Kit (Beyotime, P0028). Preparation of proximity labeling samples and streptavidin affinity pull-down The method of preparation of adjacent labeled samples is mainly referenced to Cho et al. ([149]50), with appropriate changes. Briefly, cells were cultured in 10-cm dishes. The following day, the cells were transfected with the pCDNA3.1-UltraID-NoB-EGFP plasmid. After 24 hours of protein expression, the medium was replaced, and 50 μM biotin was added. The cells were then incubated for an additional 18 hours, followed by three washes with PBS. The cells were lysed using 1 ml of RIPA lysis buffer, and the lysates were centrifuged at 12,000 rpm for 5 min at 4°C to collect the supernatant. The collected supernatant was added to streptavidin agarose beads (Beyotime, P2159) and incubated at room temperature with rotation for 1 hour. Last, the beads were washed three times by centrifugation at 600g for 5 min at 4°C using phosphate-buffered saline containing Tween-20 (PBST). Liquid chromatography–tandem mass spectrometry and data analysis Proteomic analysis was performed as previously described ([150]53). Briefly, samples collected through streptavidin affinity pull-down (20 μl of streptavidin agarose beads) were washed twice with 1 ml of NH[4]HCO[3] (50 mM) and then resuspended in a solution containing 100 μl of NH[4]HCO[3] (50 mM) and 10 μl of dithiothreitol (100 mM). The mixture was incubated at 56°C for 30 min. Subsequently, 10 μl of 300 mM iodoacetamide was added, and the samples were incubated at room temperature in the dark for 15 min. Trypsin (1:50, trypsin/lysate ratio) was then added, and digestion was carried out overnight at 37°C. On the next day, trypsin was added again, and digestion continued at 37°C for an additional 5 hours. Digests were centrifuged using a 3-kDa filter to ensure only peptides passed through. The concentrations of peptides were determined by means of a modified Lowry Protein Assay Kit (Sangon Biotech Co.). Approximately 20 μg of peptides were desalted using Pierce C18 Spin Columns (Thermo Fisher Scientific) according to the manufacturer’s directions. The peptides were analyzed using a Q Exactive HF Orbitrap MS (data are available via ProteomeXchange with identifier PXD059125). Protein identification and label-free quantification were performed using the Proteome Discoverer software (version 2.2) with default settings. Proteins with an abundance greater than 55 were considered significant. A Student’s t test (P < 0.05) was applied to identify differentially abundant proteins. Log[2] transformation was applied to the protein abundance ratios, with only proteins exhibiting a log[2] value of >1 or <−1 being deemed significant. The functions of differentially abundant proteins were retrieved from the LegioList database [151]www.uniprot.org/database/DB-0054) and UniProt ([152]www.uniprot.org/). Functional category annotations and gene essentiality data were obtained from these databases. Pathway enrichment analyses were conducted using the KEGG and Reactome platforms via the free online data analysis tool OE Biotech Tools ([153]https://cloud.oebiotech.com/#/bio/tools). Statistical analysis The data were presented as the means ± SD, derived from three independent experiments, and analyzed using GraphPad Prism version 6.01 software (San Diego, CA, USA). The significance of differences between groups was assessed using one-way analysis of variance (ANOVA) with Tukey’s correction for multiple comparisons with GraphPad Prism (version 6.01; GraphPad Software, USA), with a threshold of P < 0.05 considered to indicate statistical significance. Acknowledgments