ABSTRACT
Nucleomodulins (NMs) are bacterial nuclear-targeted effector proteins that interfere with a multitude of host cellular processes to shield pathogens from host immune responses. Recent years observed a surge in NM-related research owing to their potential role in both infectious diseases and cancer development. However, considering the complex nature of the interaction between NM and host factors, the field of “NM-disease axis” is still in the nascent phase. Thus, a comprehensive view of the known mechanisms of translocation of NMs to the host cell nucleus and mode of action, thereafter, is crucial toward deeper exploration of the “NM-disease axis.” The human gut is the major host niche to harbor bacterial cells (as part of “gut microbiota”). The current review provides an extensive collation of nucleomodulin-mediated mechanisms employed by opportunistic gut pathogens. The insights from the review would help in designing future experiments toward utilizing the NM-associated host-pathogen interaction modules in disease diagnostics and therapy.
KEYWORDS: nucleomodulins, gut pathogens, gut microbiome, nuclear localization signals, virulence factors, gene expression
INTRODUCTION
Bacterial pathogens possess a dynamic ability in adapting to highly sophisticated mechanisms to counter the complex network of immune responses produced by the host (1, 2). The success of a pathogen is governed by continual variation in the strategies employed by the bacterium in an attempt to persist intracellularly and spread infection. Some examples of such strategies include bacterial cell surface modifications, release of toxins, alterations in antigenic properties, and mimicking of host proteins (2). This review is based on one such pathogenic adaptation that involves secretion of bacterial effector molecules called nucleomodulins (NMs). The uniqueness of nucleomodulins lies in their potential to access the host cell nucleus, appositely known as the control center of the cell, unlike the widely discussed pathogenic strategies modulating host cytosolic pathways (3, 4).
The term “nucleomodulins” was coined in 2012 by Bierne and Cossart (5). As the name implies, NMs refers to bacterial molecules that have the potential to translocate to the host nucleus (“nucleo”) and modulate (“modulins”) host cell responses, which may have a long-term genetic or epigenetic effect on the host (5, 6). Most early reports on such bacterial molecules pertain to the phytopathogen, Agrobacterium tumefaciens, which is a causal agent for crown gall tumor in plants and is known to transfer and integrate bacterial plasmid DNA into the host genome (7). In the context of the human host, certain opportunistic pathogens have been reported to harbor nucleomodulins that interfere with host cellular functions, such as cell cycle processes, cell signaling pathways, and chromatin remodeling, leading to alterations in the transcriptional networks, as well as the physical state of the nuclei (3, 5, 6).
One of the commonly discussed mechanisms of nucleomodulin translocation involves mimicking the strategies employed by human nuclear-targeted proteins. This primarily includes harboring eukaryotic nuclear localization signals (NLS), which are short peptide sequences that facilitate the transport of proteins to the nucleus through the nuclear pore complex (NPC), enabling them to traverse across the nuclear envelope (8). Depending on the amino acid sequence of this motif, NLSs are classified broadly into two types, classical (cNLS) and non-classical NLS (ncNLS) (9–11). cNLS are categorized into monopartite (MP) signals, which consist of 4–8 basic amino acids like arginine (R) and lysine (K), and “bipartite” (BP) signals, which comprise two clusters of basic amino acids separated by a linker amino acid sequence. The import of NMs into the host nucleus is facilitated by “importins,” which belong to a class of proteins known as “karyopherins” (9, 10). In classical import, NLS is recognized by importin α, which binds to the cargo and presents it to importin β that carries the complex into the host nucleus. Classical signals are further classified into six classes based on their binding properties with importin α (12). NLS that do not resemble the canonical classical signals are termed as ncNLS (13, 14). One example is “PY-NLS,” which features a distinct R/K/H(X)2-5PY (where X2-5 represents any sequence consisting of 2 to 5 amino acid residues) motif located at the C-terminus. Another example includes NLS of Rex protein of human T-cell leukemia virus type 1, which comprises long stretches of arginine residues (14, 15). In both of these cases, NLS is directly recognized by importin β, bypassing the initial recognition by the adapter protein importin α, which is a characteristic of cNLS transport (14, 15). In addition to the aforementioned mimicking strategies employed by NMs, the ones lacking NLS have been reported to either diffuse through the nuclear pores (MW <50 kDa) or hijack eukaryotic proteins trafficked to the host nucleus. However, these mechanisms are just a few of the several untapped strategies employed by the pathogen that require further exploration (8, 15).
Understanding these bacterial virulence mechanisms is crucial in the context of “host-microbiome interaction.” “Microbiome” refers to the entire genetic make-up of the microbial community (or “microbiota”) that reside in a particular niche (16). Our “gut microbiota” comprises of ~1,000 resident bacterial species (along with other microbial species) with around three million genes, out of which a fraction is capable of transitioning into opportunistic pathogens (17–20). These pathogens may utilize nucleomodulin-mediated mechanisms to interfere with host pathways. In view of this, the current review aims to collate information on nucleomodulins from gut bacteria and various mechanisms employed by these proteins to manipulate a multitude of host cell signals in favor of pathogen survival. Such a comprehensive view will deepen our understanding about the capabilities of gut pathogens in controlling the host nucleus and orchestrating mechanisms that lead to cell damage. This, in turn, could potentially facilitate the design of disease management strategies to effectively combat infections by manipulating the proteins that hamper the functioning of the cell’s command center.
GUT BACTERIA AND NUCLEOMODULINS
Recent literature has demonstrated the presence of nucleomodulins in multiple gut pathogens including Salmonella typhimurium, Klebsiella pneumoniae, Helicobacter pylori, and Escherichia coli (20–23). The identification of nucleomodulins in vitro typically involves fluorescent tagging, enabling the visualization of proteins translocated to the nucleus for initial screening. Additional experimental approaches are often necessary to elucidate the specific mechanisms used by nucleomodulins to enter the host nucleus and to determine the functional consequences on cellular processes (24). Various computational tools are also available for in silico identification of cNLSs such as NLStradamus, NLS Mapper, NucImport, and NucPred (25–28). Furthermore, studies involving site-directed mutagenesis have been performed to explore the significance of NLS in the interaction of nucleomodulins with other proteins (24). Table 1 provides a summary of NLSs found in various gut pathogens. The characteristics of NLSs, the mechanism of translocation of NMs (secreted by different gut bacteria) to the host cell nucleus (schematically shown in Fig. 1), and the mode of action thereafter have been discussed in detail in the following sections.
TABLE 1.
Summary of nucleomodulins secreted by opportunistic gut pathogensa
| Pathogen | Nucleomodulin (protein name, ID, or category)b |
Mode of entering the nucleus | Method used for probing nuclear entry | StartNLS sequenceEnd | Signal type | Mode of action (reference) |
|---|---|---|---|---|---|---|
| Acinetobacter baumannii | OmpA | Using NLS | Site-directed mutagenesis | 320KTKEGRAMNRR330 | MP and cNLS | Hinders physiological state of the cell, causing cell death (29) |
| Tnp | Using NLS | Site-directed mutagenesis | 225RKRKRK230 | MP and cNLS | Suppresses E-cadherin expression by inducing DNA methylation of CpG islands in the promoter region (30) | |
| Campylobacter jejuni | Cas9 | Using NLS | Signal deletion | 44RRLARSARKRLARRKAR60 | BP and cNLS | Induces DNA damage (31, 32) |
| CdtB | Unknown | – | –c | – | Cell-cycle arrest at G2/M 50 | |
| Enterococcus faecalis | Whole bacterial cell | Unknown | Fluorescence-activated cell sorting (FACS) | – | – | Suppresses NF-κB activation in macrophages (33) |
| Escherichia coli | CdtB | Using NLS | Site-directed mutagenesis | 195REPADLEMNLTVPVRR210 | BP and cNLS | Induced DNA damage, interrupts the cell cycle (23) |
| 253RRTQISSDHFPVGVSRR269 | ||||||
| Cif | Unknown | – | – | – | Induces accumulation of the cyclin-dependent kinase inhibitors p21 and p27, causing cell cycle arrest (34, 35) | |
| EspF | 51aa domain at the N-terminus | Site-directed mutagenesis |
21SRVSSAGGTGFSVAPQAVRLTPVRVHSPFSPGSSNVNARTIFNVSSQVTS FTPS 74 |
ncNLS | EspF inflicts damage on the surrounding nucleolin, thereby affecting ribosome biogenesis (36) | |
| Tus | Using NLS | Site-directed mutagenesis | 227KLKIKRPVK235 | NLS-like sequence | Unknown | |
| Helicobacter pylori 26695 | HP0425 | Using NLS | Site-directed mutagenesis | 3KKELLKMSKKR13 | MP and cNLS | Genomic DNA degradation by DNase I-like enzymatic activity (24) |
| HP0059 | Using NLS | Site-directed mutagenesis | 188DKLKKL193 | MP and cNLS | Genomic DNA degradation by DNase I-like enzymatic activity (22) | |
| Omp18 | Using NLS | Site-directed mutagenesis | 57PKKPKRKL63 | MP and cNLS | Aids in virulence optimization for steady persistence of Hp in the host cell (37) | |
| Response regulator | Using NLS | Site-directed mutagenesis | 120KKHPLEKPLKK130 | BP and cNLS | Unknown (38) | |
| Secreted protein involved in flagellar motility | Using NLS | Site-directed mutagenesis | 66KRKRWYELFKKKPK79 | BP and cNLS | Unknown (38) | |
| UreA | Using NLS | Site-directed mutagenesis | 21KKRKEK26 | MP, cNLS | Regulates cell cycle progression by induction of hypoxia-induced factor −1α through TLR2 activation, leading to low levels of cyclin D1 (half-life) (39, 40) | |
| Klebsiella pneumoniae | HsdM | Using NLS | Site-directed mutagenesis | 7KKAKAKK13 | MP, cNLS | Methylates eukaryotic DNA, unknown mechanisms (41) |
| Listeria monocytogenes | LntA | Using NLS | In silico identification | 122IDAIKRSSEASADTEAFKKIFKEW144 | BP and cNLS | Interferes with chromatin assembly by preventing recruitment of BAHD1 to ISGs, thereby inducing their expression (42) |
| OrfX | Unknown | – | – | – | Targets RybP and reduces the oxidative capability of macrophages (43) | |
| Mycobacterium tuberculosis | Rv1988 | NLS-like sequence | Site-directed mutagenesis | 137RR139, 152RR154, and 169RRRK173 | – | Dimethylates arginine in histone H3, leading to suppression of genes implicated in inhibiting the action of macrophages (44) |
| Rv0256c | Using cNLS | Site-directed mutagenesis | 473RRRRPKIKQ481 | MP and cNLS | Inhibits nitric oxide production in infected macrophages to thrive intracellularly (45, 46) | |
| Rv2966c | NLS-like sequence, via NPM1 shuttling protein (speculated) | Site-directed mutagenesis | – | – | Methylates non-CpG distinctly CpA and CpT dinucleotides of specific gene regions (47) | |
| Rv2067c | 30 aa domain at the C-terminus | Signal deletion | – | – | Trimethylates H3K79 in free H3 histones, resulting in inhibition of caspase-dependent apoptosis (48) | |
| Rv3423.1 | Diffusion through nuclear pores (speculated) | – | – | – | Regulation of genes by acetylating histone H3 at the K9/K14 positions (49) | |
| Porphyromonas gingivalis | HRgpA | Unknown | – | – | – | Interferes with proteins implicated in cell cycle control processes (50) |
| Proteus mirabilis | Urease | Using cNLS | In silico identification | 22RRLAKGLKLNYPEAVALISCAIMEGAREGKTVA54 | BP, cNLS | Exerts pro-inflammatory effects on cells in vitro (51) |
| 373RTWQCAHKMKLQRGTLAGDSADNDNNRIKRYIAK406 | ||||||
| Salmonella enterica | SSph1 | PKN1-assisted entry, lacks cNLS | – | – | – | Interacts with PKN1 to downregulate pro-inflammatory cytokine levels by inhibition of NF-κB-dependent gene expression (52) |
| PipA GogA GtgA |
Unknown | – | – | – | Cleave nuclear transcription factors RelA and RelB to modulate the NF-κB signaling pathway and influence host transcription (53) |
BAHD1, bromo adjacent homology domain-containing 1; BP, bipartite; CdtB, cytolethal distending toxins; Cif, cycle-inhibiting factor; HRgpA, arginine-specific gingipain; ISGs, interferon-stimulated genes; LntA, Listeria nuclear targeted protein A; MP, monopartite; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLS, nuclear localization signal; NPM1, nucleophosmin; Omp, outer membrane protein; OrfX, open reading frame X; PKN1, protein kinase N1; RybP, RING1- and YY1-binding protein; SSph1, Salmonella secreted protein H1; TLR2, Toll-like receptor 2; Tnp, transposase; Tus, terminus utilization substance; TSGs, tumor suppressor genes.
In the case of Enterococcus faecalis, the whole bacterial cell acts as a nucleomodulin.
– denotes not applicable.
Fig 1.
Different strategies employed by bacterial nucleomodulins to enter the host nucleus. (A) In classical importin-mediated transport, nucleomodulins (NM) comprising classical nuclear localization signal (cNLS) are recognized by importin α. This complex is then carried via nuclear pore complex (NPC) by importin β. (B) In non-classical importin-mediated transport, NMs comprising a non-classical NLS are recognized directly by importin β, thus mediating the transport across NPC. (C) Bacterial proteins hijack eukaryotic nuclear-targeted proteins having NLS, thereby gaining entry to the nucleus. (D) NMs with molecular weight <50 kDa enter the host nucleus by passive diffusion through NPC. (E) Unexplored mechanisms.
SUPPRESSION OF CYTOKINE PRODUCTION BY SALMONELLA TYPHIMURIUM
Salmonella typhimurium is a non-typhoidal strain often associated with gastroenteritis (53). Proteins from this bacterium reported to be translocated to the nucleus include SSpH1, PipA, GtgA, and GogA (20, 53). SSpH1 is a Type III secretion system (T3SS)-mediated bacterial effector E3 ubiquitin ligase with LPX repeats (20, 54). In early reports, since SSpH1 lacks cNLS, it was hypothesized to enter the host nucleus with the help of eukaryotic protein kinase, PKN1. However, recent reports suggest the interaction of the SSph1 LRR domain with the HR1b subdomain of PKN1 in the cytoplasm, resulting in the ubiquitylation of PKN1 in vitro, thereby suppressing androgen receptor signaling (52). On another account, SspH1 enters the host cell nucleus and is known to suppress nuclear factor-kappa B (NF-κB)-dependent gene expression, resulting in downregulation of proinflammatory cytokine production (55). Another set of NMs from S. typhimurium includes GogA, GtgA, and PipA, which are homologous zinc metalloproteases that are part of the PipA family of effector proteins. These NMs proteolytically cleave transcription factors RelA and RelB, thereby deregulating NF-κB expression, resulting in attenuation of initial inflammatory responses. As a result, pathogens can persist in the system for extended durations. Sun et al. (53) demonstrated severe inflammation and increased virulence in a mouse model lacking GogA, GtgA, and PipA, thereby highlighting their role in maintaining virulence levels that do not induce inflammation (53, 56).
HOST CELL DNA METHYLATION BY KLEBSIELLA PNEUMONIAE
Klebsiella pneumoniae is a well-established opportunistic, multidrug-resistant gut pathogen, commonly implicated in hospital-acquired urinary tract infections (21). K. pneumoniae encodes HsdM, a DNA methylase and a subunit of the Type I restriction modification system, which has been experimentally shown to be translocated to the host nucleus. This translocation is facilitated by an MP cNLS sequence 7KKAKAKK13, which is recognized by the eukaryotic importin α protein. Although HsdM was not found to critically damage eukaryotic DNA in vitro (41), detailed investigation of the biological relevance of DNA methylation by HsdM would be crucial for better understanding the virulence mechanisms of K. pneumoniae.
NUCLEOMODULINS AIDING IN THE STEADY PERSISTENCE OF HELICOBACTER PYLORI
Helicobacter pylori has been implicated in around 90% of gastric cancer cases (57, 58). Cytotoxin-associated gene A (CagA) is a well-studied primary virulence factor associated with H. pylori infections. CagA, an oncoprotein involved in gastric adenocarcinomas in mammals (59), is directed to gastric epithelial cells with the help of the Type IV secretion system (T4SS). Once secreted by the bacterial cell, CagA anchors to the inner leaflet of the host cell’s plasma membrane, where it undergoes tyrosine phosphorylation, leading to a series of reactions that ultimately activate specific Wnt target genes (39, 59). While the main mechanism occurs in the cytoplasm, there have been reports of CagA being detected in the nucleus as well. However, its exact role in the nucleus remains poorly understood (60). Besides these mechanisms, two more proteins, HP0425 and HP0059, have been shown to harbor the MP cNLS sequence. These proteins with DNase I-like activity have been experimentally determined to enter the host nucleus and degrade host genomic DNA, resulting in subsequent cell death (22, 24). Further, H. pylori UreA, the structural subunit of urease, also contains a functional MP cNLS, which enables its entry into the host nucleus (38). Apart from H. pylori UreA, ureases in general have been suggested as virulent factors in various pathogenic bacteria, including Staphylococcus spp., Ureaplasma urealyticum, Corynebacterium spp., and Klebsiella spp. (40). H. pylori urease is an immunogen that is upregulated in patients with gastric cancer (61). During infection, it is delivered to the host cells via outer membrane vesicles, leading to the induction of hypoxia-inducible factor 1-alpha (HIF-1α) through TLR2 activation, thereby lowering levels of cyclin D1 and affecting cell cycle progression (62). Urease, typically a cytosolic enzyme, may also exert an influence within the host nucleus, potentially causing changes in the cell cycle beyond its primary physiological function of acid neutralization. Mutations in the UreA NLS were shown to impair the ability of the protein to enter the nucleus of cultured gastric epithelial cells, thereby causing UreA to be exclusively located in the cytoplasm (38). Lee et al. (63) demonstrated that UreA regulates a multitude of morphogenesis-related genes when present in the nucleus (63). Another bacterial urease reported to enter the host nucleus is observed in Proteus mirabilis, a microbe associated with urinary tract infections and also a commensal in the human gastrointestinal tract (64). Although P. mirabilis urease comprises two putative BP NLSs, the functionality of these NLSs has not yet been established. However, nuclear P. mirabilis urease has been reported to exert pro-inflammatory effects on the host cells in vitro (51).
Other H. pylori proteins translocated to the nucleus include outer membrane protein 18 (Omp18), transcriptional activator FlgR, and META domain-containing protein (38). Among these proteins, OMPs usually play a role in imparting virulence or assisting other virulent factors by facilitating adhesion for effective colonization inside the host (37, 65). Omp18, an antigenic protein expressed by most H. pylori strains, contains a monopartite cNLS that allows it to enter the host nucleus (38). In the nucleus, it was shown to modulate the levels of primary virulent factors, CagA and NapA, leading to a reduction in IFN-γ-mediated immune responses aimed at clearing the pathogen (37). In addition to Omp18, OMPs in H. pylori, such as SabA, OipA, and HopQ, are also recognized for their role in promoting bacterial pathogenesis. The expression of these virulence factors is regulated in response to the gastric environment (65). In-depth analysis of the aforementioned proteins may reveal additional regulatory mechanisms or novel pathways employed by H. pylori that contribute to promoting gastric metastasis.
CELL CYCLE CONTROL BY ESCHERICHIA COLI, CAMPYLOBACTER JEJUNI, AND PORPHYROMONAS GINGIVALIS NUCLEOMODULINS
Several gut bacteria such as Escherichia coli, Campylobacter jejuni, and Salmonella enterica synthesize cytolethal distending toxins (Cdts), which are known to interfere with host cell cycle processes (66). Cdts comprise three subunits, CdtA, CdtB, and CdtC. CdtA and CdtC assist in tethering the toxin to the plasma membrane of target cells (23, 66). CdtB is a homolog of eukaryotic deoxyribonuclease I that translocates to the host nucleus, causing double-strand breaks in DNA, leading to cell death (66). CdtB from E. coli, Aggregatibacter actinomycetemcomitans, and Haemophilus ducreyi has been thoroughly researched to comprehend the mechanism of its translocation to the nucleus (23). In spite of a high degree of conservation in the homologs of the CdtB subunit, the location of the NLS might differ among these proteins. Two putative NLSs have been reported in E. coli CdtB (23, 66). Although mutations in either of these NLSs did not affect the DNase activity, they impaired the protein’s ability to access the host nucleus and induce cell cycle arrest (66). C. jejuni CdtB employs unknown mechanisms distinct from typical CdtB, as observed in other organisms (67).
Cycle inhibiting factor (Cif), also a well-studied cyclomodulin in E. coli, is a protease comprising cysteine, histidine, and a glutamine catalytic triad (68). It is injected into the host cell via T3SS and is primarily known to irreversibly block cell cycle progression. Inside the host nucleus, Cif deamidates NEDD8 (neuronal precursor cell-expressed developmentally downregulated protein 8) and induces accumulation of the cyclin-dependent kinase inhibitors p21 and p27 (68). NEDD8 is a ubiquitin-like protein that activates cullin-RING E3 ubiquitin ligase (CRL) by NEDDylating the cullin subunits (68, 69). This marks the proteins for ubiquitin-based degradation by the 26S proteasome. However, in case of E. coli infection, Cif interacts with NEDD8 and obstructs the cascade, leading to ubiquitin-mediated p21 and p27 degradation, thus halting the cell cycle at the G1/S and G2/M phases (34, 35, 68). Cif lacks a cNLS signal, and the mechanism of its transport to the host nucleus has not yet been characterized.
The third E. coli effector protein, EspF, lacks a classical nuclear/nucleolar localization signal but contains a 51-amino-acid domain located at the N-terminus, which aids in targeting it to the nucleolus. Intriguingly, EspF is first accumulated in the mitochondria, which then controls the scope of EspF’s nucleolar entry. However, the regulatory facet of EspF during the course of infection has been minimally investigated and requires further examination. Once in the nucleolus, EspF inflicts damage on the most abundant protein in the vicinity, nucleolin, thereby affecting ribosome biogenesis (36).
Tus and CjeCas9 are among the other nuclear-targeted proteins from E. coli and C. jejuni, respectively (31, 70). Tus proteins are terminator proteins that halt replication by binding to terminator sequences (71). E. coli Tus comprises an NLS-like sequence, 227KLKIKRPVK235, located at the C-terminus (70). The dual functionality of the Tus protein in modulating host cell signals and facilitating infection remains to be investigated in depth. The functional role of another nucleomodulin, colibactin from E. coli, in colorectal cancer has been well elucidated (72). Although colibactin is known to enter the nucleus and induce interstrand cross-links and double-strand breaks in DNA (73), the mechanism it employs to enter the nucleus remains unknown. C. jejuni protein CjeCas9 nuclease enters host cells via outer membrane vesicles and subsequently reaches the nucleus with the help of a BP cNLS, 44RRLARSARKRLARRKAR60, where it induces cell damage by degrading chromatin (31, 32).
Another nucleomodulin known to interfere with cell cycle processes is protease HRgpA (arginine-specific gingipain) from Porphyromonas gingivalis (50). Although P. gingivalis is an oral anaerobe that contributes to the pathogenesis of periodontitis, it is frequently observed to be translocated to the gut in patients with pancreatic cancer and rheumatoid arthritis (74–76). HRgpA has been reported to effectively translocate to the host cell nucleus in vitro, but its mechanism of translocation remains unclear (50).
CHROMATIN ASSEMBLY INTERFERENCE BY LISTERIA MONOCYTOGENES
Listeria monocytogenes is an opportunistic pathogen that colonizes the gastrointestinal tract during listeriosis, an infectious foodborne disease (77). L. monocytogenes produces several virulence factors including NMs such as LntA, OrfX, and InlP (42, 43, 78). The entry of LntA into the host nucleus is speculated to be facilitated by a putative BP cNLS, determined in silico. LntA targets bromo adjacent homology domain containing protein 1 (BAHD1), a transcriptional repressor implicated in the formation of heterochromatin (42, 79). It interferes with chromatin assembly by preventing recruitment of BAHD1 to interferon-stimulated genes (ISGs), thereby inducing their expression, resulting in colonization of L. monocytogenes by modulating IFN- γ-mediated immune response (42). Downregulation of BAHD1 has been shown to decrease the chance of survival in the case of lung cancer patients, suggesting its importance during the infection (42, 80). Another nucleomodulin from L. monocytogenes is OrfX, which lacks cNLS, and the mechanisms employed for facilitating its nuclear entry are yet to be characterized (43). This nucleomodulin targets host nuclear zinc finger protein RybP (RING1- and YY1-binding protein), which is one of the critical proteins involved in developmental as well as proapoptotic pathways (43, 81). RybP is also known to be oncogenic in nature as it is involved in averting proteasome-mediated ubiquitination of tumor suppressor protein p53 by interacting with E3 ubiquitin-protein ligase MDM2 (43, 82). OrfX is positively regulated by PrfA, which is overexpressed during L. monocytogenes infection and is reported to activate other vital virulence proteins. Overall, the interaction of OrfX and RybP leads to dysfunctionality of macrophages in initiating oxidative burst, thus aiding in the intracellular survival of the bacteria (43). Additionally, the third nucleomodulin, Internalin P (InlP) from L. monocytogenes, targets nuclear speckles, which are dynamic structures constituting pre-mRNA splicing components (83). The specific mechanism by which InlP translocates to the nucleus remains unclear; however, once in the host nucleus, it specifically targets RBM5 (RNA-binding motif protein 5), an mRNA splicing regulator involved in cell cycle regulation and apoptosis (78, 83, 84).
INDUCTION OF DNA METHYLATION BY ACINETOBACTER BAUMANNII
Acinetobacter baumannii is an opportunistic pathogen primarily involved in nosocomial infections where it colonizes the gut and often develops antimicrobial resistance (85). While 15 proteins from A. baumannii have been experimentally validated to target the host nucleus, the mechanism of action has been clearly elucidated only for two of these proteins, transposase (Tnp) and outer membrane protein A (AbOmpA) (29, 30). A. baumannii entry to the cytoplasm of host cells is facilitated by outer membrane vesicles. Once in the cytoplasm, Tnp is delivered to the host nucleus via MP cNLS 225RKRKRK230, which then contributes to epigenetic modifications in the host cell, leading to the downregulation of E-cadherin expression (29, 30, 85, 86). Tnp suppresses E-cadherin expression by inducing DNA methylation in the CpG islands of the promoter region of the gene (30). E-cadherin is a tumor suppressor gene, the loss of which, along with that of some other proteins, has been implicated in the initiation of epithelial mesenchymal transition (EMT) in cancer, a process that drives tumor metastasis (87–89). Another A. baumannii protein, OmpA, which is involved in apoptosis of epithelial cells, also targets the nuclear compartment of the host cell using an MP cNLS 320KTKEGRAMNRR330. Choi et al. (29) through site-directed mutagenesis studies demonstrated the significance of two lysine residues in the NLS that enable this nucleomodulin to enter the host nucleus (29, 86). Interestingly, OmpA has also been reported to translocate to the mitochondria, and this colocalization is suggested to be crucial for hindering the physiological state of the cell, eventually resulting in cell death (29).
MASTER EPIGENETIC REGULATOR MYCOBACTERIUM TUBERCULOSIS
Mycobacterium tuberculosis is a pathogen known to exhibit a range of evading mechanisms by hijacking host immune responses. Although a respiratory pathogen, it can translocate to the gut, particularly in the ileocecal region, leading to gastrointestinal tuberculosis (90). Rv1988, Rv2067c, Rv3423.1, Rv2966c, and Rv0256c are M. tuberculosis nucleomodulins that directly interfere with immune defense mechanisms by manipulating the host’s epigenetic landscape (44, 47–49). These mechanisms have been schematically depicted in Fig. 2. Rv1988 is a methyltransferase secreted via the Tat-secretion pathway in virulent mycobacterial strains. It dimethylates arginine in histone H3 (H3R42), resulting in suppression of genes encoding NADPH oxidases (NOX1 and NOX4) and nitric oxide synthase (NOS2) (44). This allows mycobacteria to overcome the initial line of defense, replicate effectively within cells, and spread the infection (44, 91). Yaseen et al. (44) demonstrated that the nuclear targeting of Rv1988 is facilitated by an arginine-rich region spanning amino acids from 137 to 179 (137RR139, 152RR154, and 169RRRK173) located at the C-terminus (44). Further, the study highlights the propensity of Rv1988 toward methylation of non-tail core histone H3-arginine H3R42, which is usually not a target for methylation by mammalian histone methyltransferases (44).
Fig 2.

Nucleomodulins secreted by M. tuberculosis in phagosome escape into the cytosol followed by translocation to the nucleus. Once inside the macrophage nucleus, (A) Rv0256c binds to the iNOS promoter and inhibits iNOS transcription, resulting in reduced nitric oxide (NO) production. (B) Rv3423.1 acetylates H3 histone, leading to alteration of anti-inflammatory responses. (C) Rv1988 methylates the arginine residue at the 42 position of histone H3, thereby suppressing the expression of genes such as NOX1, NOX4, and nitric oxide synthase 2 (NOS2), which are involved in the production of reactive oxygen species (ROS). (D) Rv2966 methylates cytosine residues of histones H3 and H4 in a non-CpG context, resulting in downregulation of genes responsible for the initial immune response. (E) Rv2067 methylates nuclear-targeted non-nucleosomal H3 histone in cytosol as well as in the nucleus, thus downregulating the epigenetic activity of human methyltransferase DOT1L. This results in upregulation of SESTRIN, TMTC1, and NLRC3 and subsequent inhibition of caspase-8-dependent apoptosis.
Rv2067c is another methyltransferase, which enters the nucleus with the help of 30 amino acid residues located in the C-terminus (48). It is capable of methylating free, non-nucleosomal histone H3 at the K79 position (H3K79) in both cytoplasmic as well as nuclear environments of the macrophages. Interestingly, Rv2067c suppresses the expression of the host methyltransferase DOT1L (Disruptor of Telomeric silencing 1-like), which is also directed toward the same methylation site as Rv2067c but in a nucleosomal H3 (48). The proclivity of both methyltransferases to target the same site in H3 varies depending on the nucleosomal context due to significant structural differences between the two methyltransferases. Rv2067 is a homodimer that contains N-terminal SAM-binding catalytic domain (CD), a dimerization domain (DD), and a C-terminal domain (CTD), whereas DOT1L is a monomer with distinctly different domains from Rv2067c, except for the shared Class I MTase fold (48). Furthermore, the binding of DOT1L induces a conformational change in the H3K79 loop, moving H3K79 from an inaccessible position into the active site of DOT1L, thereby aiding the process of methylation. This epigenetic marker upregulates the expression of genes encoding proteins such as TMTC1, SESTRIN3, and NLRC3, collectively shielding pathogens from host immune responses (48).
A third methyltransferase, Rv2966c, was found in the nucleus of THP1 cells in vivo and was further determined to bind and methylate non-CpG (distinctly CpA and CpT) dinucleotides (47). A stretch of 20 amino acids located in the C-terminal region, comprising three arginine (at positions 171, 177, and 183) and one threonine residue, was suggested to be the signal sequence of Rv2966c. In addition to arginine, the mutation in T176 also affected the nuclear targeting of the protein. Based on the interactions between nucleophosmin (NPM1) and Rv2966 through in vitro experiments, Sharma et al. (47) suggested the potential role of the NPM1 protein in the nuclear targeting of Rv2966. Post-translational modifications, such as phosphorylation, by PknB were reported to increase the DNA binding and methylating activity of Rv2966c. Rv2966c was also found to bind to specific gene regions including one within H2AFY2, a gene that codes for macro histone and G protein-coupled receptor kinase 5 (GRK5). Further, in support of “Rv2966-GRK5”-mediated pathogenesis, GRK5 has been reported to be significantly downregulated in Mycobacterium-infected THP1 macrophages and has previously been shown to interact with DNA and proteins other than GPCR in the nucleus (47).
Rv3423.1 is another mycobacterial epigenetic regulator of gene expression during infection. Rv3423.1 is a histone acetyltransferase and is reported to acetylate histone H3 at the K9/K14 positions. Given the small size of this protein (8.4 kDa) and lack of functional NLS, it is speculated that it may diffuse through nuclear pores or hijack eukaryotic nuclear-targeted protein to gain access to the host nucleus (49). The role of this protein once it is in the nucleus is not yet well-elucidated. However, given the nature of this enzyme, it is suggested that it may be involved in escaping initial host immune responses, thereby prolonging the bacterial intracellular survival period inside macrophages (49, 92).
Furthermore, another nucleomodulin secreted by M. tuberculosis is Rv0256c (PPE2), which comprises proline-glutamate (PE) and proline-proline-glutamate (PPE) residues. The presence of functional MP NLS located at the C-terminal facilitates nuclear entry of PPE2 mediated via the classical importin α/β pathway, mimicking eukaryotic protein nuclear transport. Once within the nucleus, PPE2 inhibits nitric oxide production in infected macrophages as it binds to the iNOS promoter and leads to masking the GATA-1 binding sites. This inhibition renders the macrophages dysfunctional, thereby conferring an intracellular survival advantage to mycobacteria (45, 46).
DISCUSSION
The human gut is home to microbial communities important for maintenance of a balanced environment. An imbalance in these communities often leads to a conducive environment for the colonization of pathogens and the development of disease conditions. Studies have shown that this dysbiosis in the microbiome is associated with modulation in host gene expression, cell signaling pathways, chromatin dynamics, and even epigenetic regulation. Bacteria can utilize multiple ways to affect the host homeostasis, including utilizing their metabolites and membrane proteins to evade host immune response and influence regulatory and signaling pathways. In addition, bacteria utilize various effector proteins to reprogram host cells using different mechanisms of action. One such class of proteins includes nucleomodulins (NMs), which attack the most crucial eukaryotic cell organelle, the nucleus. Bacterial NMs have evolved to possess classical signal sequences that facilitate nuclear localization in the host cells, thereby mimicking eukaryotic nuclear proteins. Translocation of these NMs into the host cell nucleus has been experimentally validated to induce a multitude of cell cycle alterations, including epigenetic rewiring, that aids in establishing a compatible niche for the pathogen to replicate and spread. As the functioning of NMs involves an interaction with an array of host proteins in the gut environment, NMs play a crucial role in shaping the dynamics of the host-microbiome interaction. The current review presents a comprehensive collation of NM-dependent strategies, primarily employed by opportunistic gut pathogens, which would help in designing future experiments toward gaining better insights into the host-pathogen (as well as host-microbiome) interaction. In addition to classical NLS-dependent translocation, the present review discusses various other nuclear translocation mechanisms reported in recent studies.
Apart from the classical and non-classical NMs discussed in the current review, Enterococcus faecalis presents a distinct case wherein the whole bacterial cells are congregated in the perinuclear region surrounding the nucleus (33). E. faecalis is a common opportunistic pathogen, which can cause antibiotic-resistant infections and has also been frequently linked to colorectal cancer (93, 94). E. faecalis was found to replicate intracellularly in both immune and non-immune cells in vitro. The initial entry of the pathogen into the host cells is facilitated by macropinocytosis, followed by the downregulation of the GTPase Rab7. This process enables the bacteria to negatively regulate endosome-lysosome fusion and replicate within the cells, thereby prolonging the infection period (33).
Experimental identification of nucleomodulins is usually performed by tracking their nuclear localization using fluorescence-based techniques, the resolution of which depends on the expression levels. Such identification can be challenging during certain stages of infection as NM expression is dominated by the expression of primary virulent factors. For instance, as previously discussed in the case of H. pylori, the nucleomodulin Omp18 regulates the expression of primary virulence factors CagA and NapA, thus escaping early host inflammatory responses aimed at eliminating the pathogen (59, 60). Therefore, it is essential to monitor the expression levels of both nucleomodulins and primary virulence factors over the course of infection, toward evaluating the complex network of these proteins driving the pathogenesis. The comprehensive collection of nucleomodulins with signal sequence information, provided in this review, can help in building robust methods capable of predicting diverse categories of bacterial NMs.
Given the ability of bacterial nucleomodulins to interact with host cell DNA and alter nuclear functions, including gene expression, recent literature suggests their diverse roles in exacerbation of various infectious diseases and cancer. As mentioned earlier, NMs from H. pylori and E. coli have been reported to be upregulated in cancer patients (61, 62, 72, 95). Further, one of the NMs from E. coli, colibactin, was found to induce specific mutation at the DNA damage site in infected human colorectal cancer cells (73, 95). Overall, involvement of bacterial infection in cancer pathophysiology is being increasingly acknowledged. Nucleomodulins, being a suspected key driver of the “bacterial infection-cancer axis,” demands focused studies toward their potential utilization in diagnostic and therapeutic regimes. Therefore, accounting for both realms of host physiology and interacting microbiome is important for improving cancer therapy as well as prognosis.
ACKNOWLEDGMENTS
We acknowledge Dr. Sharmila Mande and Ms. Vatsala Pokhrel for their valuable input and suggestions.
AFTER EPUB
[This article was published on 18 July 2025 with typographical errors in the Introduction and Discussion. The errors were corrected in the current version, posted on 23 July 2025.]
Contributor Information
Chandrani Bose, Email: chandrani.das@tcs.com.
Swadha Anand, Email: swadha.anand@tcs.com.
Issac Cann, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.
REFERENCES
- 1. Felden B, Cattoir V. 2018. Bacterial adaptation to antibiotics through regulatory RNAs. Antimicrob Agents Chemother 62:e02503-17. doi: 10.1128/AAC.02503-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Finlay BB, McFadden G. 2006. Anti-immunology: evasion of the host immune system by bacterial and viral pathogens. Cell 124:767–782. doi: 10.1016/j.cell.2006.01.034 [DOI] [PubMed] [Google Scholar]
- 3. Ribet D, Cossart P. 2015. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect 17:173–183. doi: 10.1016/j.micinf.2015.01.004 [DOI] [PubMed] [Google Scholar]
- 4. Cooper GM. 2000. Chapter 8, The nucleus. In The cell: a molecular approach, 2nd ed. Sinauer Associates, Sunderland (MA). [Google Scholar]
- 5. Bierne H, Cossart P. 2012. When bacteria target the nucleus: the emerging family of nucleomodulins. Cell Microbiol 14:622–633. doi: 10.1111/j.1462-5822.2012.01758.x [DOI] [PubMed] [Google Scholar]
- 6. Hanford HE, Von Dwingelo J, Abu Kwaik Y. 2021. Bacterial nucleomodulins: a coevolutionary adaptation to the eukaryotic command center. PLoS Pathog 17:e1009184. doi: 10.1371/journal.ppat.1009184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Gohlke J, Deeken R. 2014. Plant responses to Agrobacterium tumefaciens and crown gall development. Front Plant Sci 5:155. doi: 10.3389/fpls.2014.00155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Cooper GM. 2000. The nuclear envelope and traffic between the nucleus and cytoplasm. In The cell: a molecular approach, 2nd ed. Sunderland (MA): Sinauer Associates. [Google Scholar]
- 9. Lu J, Wu T, Zhang B, Liu S, Song W, Qiao J, Ruan H. 2021. Types of nuclear localization signals and mechanisms of protein import into the nucleus. Cell Commun Signal 19:60. doi: 10.1186/s12964-021-00741-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Lange A, McLane LM, Mills RE, Devine SE, Corbett AH. 2010. Expanding the definition of the classical bipartite nuclear localization signal. Traffic 11:311–323. doi: 10.1111/j.1600-0854.2009.01028.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. McLane LM, Corbett AH. 2009. Nuclear localization signals and human disease. IUBMB Life 61:697–706. doi: 10.1002/iub.194 [DOI] [PubMed] [Google Scholar]
- 12. Kosugi S, Hasebe M, Matsumura N, Takashima H, Miyamoto-Sato E, Tomita M, Yanagawa H. 2009. Six classes of nuclear localization signals specific to different binding grooves of importin alpha. J Biol Chem 284:478–485. doi: 10.1074/jbc.M807017200 [DOI] [PubMed] [Google Scholar]
- 13. Cingolani G, Bednenko J, Gillespie MT, Gerace L. 2002. Molecular basis for the recognition of a nonclassical nuclear localization signal by importin beta. Mol Cell 10:1345–1353. doi: 10.1016/s1097-2765(02)00727-x [DOI] [PubMed] [Google Scholar]
- 14. Palmeri D, Malim MH. 1999. Importin beta can mediate the nuclear import of an arginine-rich nuclear localization signal in the absence of importin alpha. Mol Cell Biol 19:1218–1225. doi: 10.1128/MCB.19.2.1218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Görlich D, Kutay U. 1999. Transport between the cell nucleus and the cytoplasm. Annu Rev Cell Dev Biol 15:607–660. doi: 10.1146/annurev.cellbio.15.1.607 [DOI] [PubMed] [Google Scholar]
- 16. Liu X. 2016. Microbiome. Yale J Biol Med 89:275–276. doi: 10.1353/tfr.2016.0197 [DOI] [Google Scholar]
- 17. Quigley EMM. 2013. Gut bacteria in health and disease. Gastroenterol Hepatol 9:560–569. [PMC free article] [PubMed] [Google Scholar]
- 18. Brown SP, Cornforth DM, Mideo N. 2012. Evolution of virulence in opportunistic pathogens: generalism, plasticity, and control. Trends Microbiol 20:336–342. doi: 10.1016/j.tim.2012.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Rajilić-Stojanović M, de Vos WM. 2014. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 38:996–1047. doi: 10.1111/1574-6976.12075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Cook M, Delbecq SP, Schweppe TP, Guttman M, Klevit RE, Brzovic PS. 2019. The ubiquitin ligase SspH1 from Salmonella uses a modular and dynamic E3 domain to catalyze substrate ubiquitylation. J Biol Chem 294:783–793. doi: 10.1074/jbc.RA118.004247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Struve C, Krogfelt KA. 2004. Pathogenic potential of environmental Klebsiella pneumoniae isolates. Environ Microbiol 6:584–590. doi: 10.1111/j.1462-2920.2004.00590.x [DOI] [PubMed] [Google Scholar]
- 22. Kwon YC, Kim S, Lee YS, Lee JC, Cho MJ, Lee WK, Kang HL, Song JY, Baik SC, Ro HS. 2016. Novel nuclear targeting coiled-coil protein of Helicobacter pylori showing Ca2+-independent, Mg2+-dependent DNase I activity. J Microbiol Seoul Korea 54:387–395. doi: 10.1007/s12275-016-5631-9 [DOI] [PubMed] [Google Scholar]
- 23. McSweeney LA, Dreyfus LA. 2004. Nuclear localization of the Escherichia coli cytolethal distending toxin CdtB subunit. Cell Microbiol 6:447–458. doi: 10.1111/j.1462-5822.2004.00373.x [DOI] [PubMed] [Google Scholar]
- 24. Kim JM, Choe MH, Asaithambi K, Song JY, Lee YS, Lee JC, Seo JH, Kang HL, Lee KH, Lee WK, Cho MJ, Rhee KH, Youn HS, Baik SC. 2016. Helicobacter pylori HP0425 targets the nucleus with DNase I-like activity. Helicobacter 21:218–225. doi: 10.1111/hel.12271 [DOI] [PubMed] [Google Scholar]
- 25. Nguyen Ba AN, Pogoutse A, Provart N, Moses AM. 2009. NLStradamus: a simple Hidden Markov Model for nuclear localization signal prediction. BMC Bioinformatics 10:202. doi: 10.1186/1471-2105-10-202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Kosugi S, Hasebe M, Tomita M, Yanagawa H. 2009. Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proc Natl Acad Sci USA 106:10171–10176. doi: 10.1073/pnas.0900604106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Mehdi AM, Sehgal MSB, Kobe B, Bailey TL, Bodén M. 2011. A probabilistic model of nuclear import of proteins. Bioinformatics 27:1239–1246. doi: 10.1093/bioinformatics/btr121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Brameier M, Krings A, MacCallum RM. 2007. NucPred—predicting nuclear localization of proteins. Bioinformatics 23:1159–1160. doi: 10.1093/bioinformatics/btm066 [DOI] [PubMed] [Google Scholar]
- 29. Choi CH, Hyun SH, Lee JY, Lee JS, Lee YS, Kim SA, Chae JP, Yoo SM, Lee JC. 2008. Acinetobacter baumannii outer membrane protein A targets the nucleus and induces cytotoxicity. Cell Microbiol 10:309–319. doi: 10.1111/j.1462-5822.2007.01041.x [DOI] [PubMed] [Google Scholar]
- 30. Moon DC, Choi CH, Lee SM, Lee JH, Kim SI, Kim DS, Lee JC. 2012. Nuclear translocation of Acinetobacter baumannii transposase induces DNA methylation of CpG regions in the promoters of E-cadherin gene. PLoS ONE 7:e38974. doi: 10.1371/journal.pone.0038974 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Saha C, Mohanraju P, Stubbs A, Dugar G, Hoogstrate Y, Kremers G-J, van Cappellen WA, Horst-Kreft D, Laffeber C, Lebbink JHG, Bruens S, Gaskin D, Beerens D, Klunder M, Joosten R, Demmers JAA, van Gent D, Mouton JW, van der Spek PJ, van der Oost J, van Baarlen P, Louwen R. 2020. Guide-free Cas9 from pathogenic Campylobacter jejuni bacteria causes severe damage to DNA. Sci Adv 6:eaaz4849. doi: 10.1126/sciadv.aaz4849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Saha C, Horst-Kreft D, Kross I, van der Spek PJ, Louwen R, van Baarlen P. 2020. Campylobacter jejuni Cas9 modulates the transcriptome in Caco-2 intestinal epithelial cells. Genes (Basel) 11:1193. doi: 10.3390/genes11101193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. da Silva RAG, Tay WH, Ho FK, Tanoto FR, Chong KKL, Choo PY, Ludwig A, Kline KA. 2022. Enterococcus faecalis alters endo-lysosomal trafficking to replicate and persist within mammalian cells. PLoS Pathog 18:e1010434. doi: 10.1371/journal.ppat.1010434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Taieb F, Nougayrède JP, Watrin C, Samba-Louaka A, Oswald E. 2006. Escherichia coli cyclomodulin Cif induces G2 arrest of the host cell cycle without activation of the DNA-damage checkpoint-signalling pathway. Cell Microbiol 8:1910–1921. doi: 10.1111/j.1462-5822.2006.00757.x [DOI] [PubMed] [Google Scholar]
- 35. Taieb F, Nougayrède JP, Oswald E. 2011. Cycle inhibiting factors (Cifs): cyclomodulins that usurp the ubiquitin-dependent degradation pathway of host cells. Toxins (Basel) 3:356–368. doi: 10.3390/toxins3040356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Dean P, Scott JA, Knox AA, Quitard S, Watkins NJ, Kenny B. 2010. The enteropathogenic E. coli effector EspF targets and disrupts the nucleolus by a process regulated by mitochondrial dysfunction. PLoS Pathog 6:e1000961. doi: 10.1371/journal.ppat.1000961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Shan Y, Lu X, Han Y, Li X, Wang X, Shao C, Wang L, Liu Z, Tang W, Sun Y, Jia J. 2015. Helicobacter pylori outer membrane protein 18 (Hp1125) is involved in persistent colonization by evading interferon-γ signaling. Biomed Res Int 2015:571280. doi: 10.1155/2015/571280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lee JH, Jun SH, Baik SC, Kim DR, Park JY, Lee YS, Choi CH, Lee JC. 2012. Prediction and screening of nuclear targeting proteins with nuclear localization signals in Helicobacter pylori. J Microbiol Methods 91:490–496. doi: 10.1016/j.mimet.2012.10.004 [DOI] [PubMed] [Google Scholar]
- 39. Noto JM, Peek RM. 2012. The Helicobacter pylori cag pathogenicity island. Methods Mol Biol 921:41–50. doi: 10.1007/978-1-62703-005-2_7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Konieczna I, Zarnowiec P, Kwinkowski M, Kolesinska B, Fraczyk J, Kaminski Z, Kaca W. 2012. Bacterial urease and its role in long-lasting human diseases. Curr Protein Pept Sci 13:789–806. doi: 10.2174/138920312804871094 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Lee JC, Kim DS, Moon DC, Lee JH, Kim MJ, Lee SM, Lee YS, Kang S-W, Lee EJ, Kang SS, Lee E, Hyun SH. 2009. Prediction of bacterial proteins carrying a nuclear localization signal and nuclear targeting of HsdM from Klebsiella pneumoniae. J Microbiol 47:641–645. doi: 10.1007/s12275-009-0217-4 [DOI] [PubMed] [Google Scholar]
- 42. Lebreton A, Job V, Ragon M, Le Monnier A, Dessen A, Cossart P, Bierne H. 2014. Structural basis for the inhibition of the chromatin repressor BAHD1 by the bacterial nucleomodulin LntA. mBio 5:e00775-13. doi: 10.1128/mBio.00775-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Bossuet-Greif N, Vignard J, Taieb F, Mirey G, Dubois D, Petit C, Oswald E, Nougayrède J-P. 2018. The colibactin genotoxin generates DNA interstrand cross-links in infected cells. mBio 9. doi: 10.1128/mBio.02393-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Yaseen I, Kaur P, Nandicoori VK, Khosla S. 2015. Mycobacteria modulate host epigenetic machinery by Rv1988 methylation of a non-tail arginine of histone H3. Nat Commun 6:8922. doi: 10.1038/ncomms9922 [DOI] [PubMed] [Google Scholar]
- 45. Bisht MK, Pal R, Dahiya P, Naz S, Sanyal P, Nandicoori VK, Ghosh S, Mukhopadhyay S. 2023. The PPE2 protein of Mycobacterium tuberculosis is secreted during infection and facilitates mycobacterial survival inside the host. Tuberculosis (Edinb) 143:102421. doi: 10.1016/j.tube.2023.102421 [DOI] [PubMed] [Google Scholar]
- 46. Pal R, Ghosh S, Mukhopadhyay S. 2021. Moonlighting by PPE2 protein: focus on mycobacterial virulence. J Immunol 207:2393–2397. doi: 10.4049/jimmunol.2100212 [DOI] [PubMed] [Google Scholar]
- 47. Sharma G, Upadhyay S, Srilalitha M, Nandicoori VK, Khosla S. 2015. The interaction of mycobacterial protein Rv2966c with host chromatin is mediated through non-CpG methylation and histone H3/H4 binding. Nucleic Acids Res 43:3922–3937. doi: 10.1093/nar/gkv261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Singh PR, Dadireddy V, Udupa S, Kalladi SM, Shee S, Khosla S, Rajmani RS, Singh A, Ramakumar S, Nagaraja V. 2023. The Mycobacterium tuberculosis methyltransferase Rv2067c manipulates host epigenetic programming to promote its own survival. Nat Commun 14:8497. doi: 10.1038/s41467-023-43940-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Jose L, Ramachandran R, Bhagavat R, Gomez RL, Chandran A, Raghunandanan S, Omkumar RV, Chandra N, Mundayoor S, Kumar RA. 2016. Hypothetical protein Rv3423.1 of Mycobacterium tuberculosis is a histone acetyltransferase. FEBS J 283:265–281. doi: 10.1111/febs.13566 [DOI] [PubMed] [Google Scholar]
- 50. Scragg MA, Alsam A, Rangarajan M, Slaney JM, Shepherd P, Williams DM, Curtis MA. 2002. Nuclear targeting of porphyromonas gingivalis W50 protease in epithelial cells. Infect Immun 70:5740–5750. doi: 10.1128/IAI.70.10.5740-5750.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Grahl MVC, Uberti AF, Broll V, Bacaicoa-Caruso P, Meirelles EF, Carlini CR. 2021. Proteus mirabilis urease: unsuspected non-enzymatic properties relevant to pathogenicity. Int J Mol Sci 22:7205. doi: 10.3390/ijms22137205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Keszei AFA, Tang X, McCormick C, Zeqiraj E, Rohde JR, Tyers M, Sicheri F. 2014. Structure of an SspH1-PKN1 complex reveals the basis for host substrate recognition and mechanism of activation for a bacterial E3 ubiquitin ligase. Mol Cell Biol 34:362–373. doi: 10.1128/MCB.01360-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Sun H, Kamanova J, Lara-Tejero M, Galán JE. 2016. A family of Salmonella type III secretion effector proteins selectively targets the NF-κB signaling pathway to preserve host homeostasis. PLoS Pathog 12:e1005484. doi: 10.1371/journal.ppat.1005484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Norkowski S, Schmidt MA, Rüter C. 2018. The species-spanning family of LPX-motif harbouring effector proteins. Cell Microbiol 20:e12945. doi: 10.1111/cmi.12945 [DOI] [PubMed] [Google Scholar]
- 55. Maculins T, Fiskin E, Bhogaraju S, Dikic I. 2016. Bacteria-host relationship: ubiquitin ligases as weapons of invasion. Cell Res 26:499–510. doi: 10.1038/cr.2016.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Takemura M, Haneda T, Idei H, Miki T, Okada N. 2021. A Salmonella type III effector, PipA, works in a different manner than the PipA family effectors GogA and GtgA. PLoS One 16:e0248975. doi: 10.1371/journal.pone.0248975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Hansson LE, Engstrand L, Nyrén O, Evans DJ, Lindgren A, Bergström R, Andersson B, Athlin L, Bendtsen O, Tracz P. 1993. Helicobacter pylori infection: independent risk indicator of gastric adenocarcinoma. Gastroenterology 105:1098–1103. doi: 10.1016/0016-5085(93)90954-b [DOI] [PubMed] [Google Scholar]
- 58. Schulz C, Schütte K, Mayerle J, Malfertheiner P. 2019. The role of the gastric bacterial microbiome in gastric cancer: Helicobacter pylori and beyond. Therap Adv Gastroenterol 12:1756284819894062. doi: 10.1177/1756284819894062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Hatakeyama M. 2014. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe 15:306–316. doi: 10.1016/j.chom.2014.02.008 [DOI] [PubMed] [Google Scholar]
- 60. Uchida T, Kanada R, Tsukamoto Y, Hijiya N, Matsuura K, Yano S, Yokoyama S, Kishida T, Kodama M, Murakami K, Fujioka T, Moriyama M. 2007. Immunohistochemical diagnosis of the cagA-gene genotype of Helicobacter pylori with anti-East Asian CagA-specific antibody. Cancer Sci 98:521–528. doi: 10.1111/j.1349-7006.2007.00415.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Wu CH, Huang MY, Yeh CS, Wang JY, Cheng TL, Lin SR. 2007. Overexpression of Helicobacter pylori-associated urease mRNAs in human gastric cancer. DNA Cell Biol 26:641–648. doi: 10.1089/dna.2007.0599 [DOI] [PubMed] [Google Scholar]
- 62. Valenzuela-Valderrama M, Cerda-Opazo P, Backert S, González MF, Carrasco-Véliz N, Jorquera-Cordero C, Wehinger S, Canales J, Bravo D, Quest AFG. 2019. The Helicobacter pylori urease virulence factor is required for the induction of hypoxia-induced factor-1α in gastric cells. Cancers (Basel) 11:799. doi: 10.3390/cancers11060799 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Lee JH, Jun SH, Kim JM, Baik SC, Lee JC. 2015. Morphological changes in human gastric epithelial cells induced by nuclear targeting of Helicobacter pylori urease subunit A. J Microbiol 53:406–414. doi: 10.1007/s12275-015-5085-5 [DOI] [PubMed] [Google Scholar]
- 64. Hamilton AL, Kamm MA, Ng SC, Morrison M. 2018. Proteus spp. as putative gastrointestinal pathogens. Clin Microbiol Rev 31:e00085-17. doi: 10.1128/CMR.00085-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Xu C, Soyfoo DM, Wu Y, Xu S. 2020. Virulence of Helicobacter pylori outer membrane proteins: an updated review. Eur J Clin Microbiol Infect Dis 39:1821–1830. doi: 10.1007/s10096-020-03948-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Guerra L, Cortes-Bratti X, Guidi R, Frisan T. 2011. The biology of the cytolethal distending toxins. Toxins (Basel) 3:172–190. doi: 10.3390/toxins3030172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lai CK, Chen YA, Lin CJ, Lin HJ, Kao MC, Huang MZ, Lin YH, Chiang-Ni C, Chen CJ, Lo UG, Lin LC, Lin H, Hsieh JT, Lai CH. 2016. Molecular mechanisms and potential clinical applications of Campylobacter jejuni cytolethal distending toxin. Front Cell Infect Microbiol 6:9. doi: 10.3389/fcimb.2016.00009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Jubelin G, Taieb F, Duda DM, Hsu Y, Samba-Louaka A, Nobe R, Penary M, Watrin C, Nougayrède JP, Schulman BA, Stebbins CE, Oswald E. 2010. Pathogenic bacteria target NEDD8-conjugated cullins to hijack host-cell signaling pathways. PLoS Pathog 6:e1001128. doi: 10.1371/journal.ppat.1001128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kamitani T, Kito K, Nguyen HP, Yeh ETH. 1997. Characterization of NEDD8, a developmentally down-regulated ubiquitin-like protein. J Biol Chem 272:28557–28562. doi: 10.1074/jbc.272.45.28557 [DOI] [PubMed] [Google Scholar]
- 70. Kaczmarczyk SJ, Sitaraman K, Hill T, Hartley JL, Chatterjee DK. 2010. Tus, an E. coli protein, contains mammalian nuclear targeting and exporting signals. PLoS One 5:e8889. doi: 10.1371/journal.pone.0008889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Neylon C, Kralicek AV, Hill TM, Dixon NE. 2005. Replication termination in Escherichia coli: structure and antihelicase activity of the Tus-Ter complex. Microbiol Mol Biol Rev 69:501–526. doi: 10.1128/MMBR.69.3.501-526.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Dubinsky V, Dotan I, Gophna U. 2020. Carriage of colibactin-producing bacteria and colorectal cancer risk. Trends Microbiol 28:874–876. doi: 10.1016/j.tim.2020.05.015 [DOI] [PubMed] [Google Scholar]
- 73. Bossuet-Greif N, Vignard J, Taieb F, Mirey G, Dubois D, Petit C, Oswald E, Nougayrède JP. 2018. The colibactin genotoxin generates DNA interstrand cross-links in infected cells. mBio 9:e02393-17. doi: 10.1128/mBio.02393-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Park SY, Hwang BO, Lim M, Ok SH, Lee SK, Chun KS, Park KK, Hu Y, Chung WY, Song NY. 2021. Oral-Gut microbiome axis in gastrointestinal disease and cancer. Cancers (Basel) 13:2124. doi: 10.3390/cancers13092124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Flak MB, Colas RA, Muñoz-Atienza E, Curtis MA, Dalli J, Pitzalis C. 2022. Inflammatory arthritis disrupts gut resolution mechanisms, promoting barrier breakdown by Porphyromonas gingivalis. JCI Insight 7:e165600. doi: 10.1172/jci.insight.165600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Chung M, Zhao N, Meier R, Koestler DC, Wu G, de Castillo E, Paster BJ, Charpentier K, Izard J, Kelsey KT, Michaud DS. 2021. Comparisons of oral, intestinal, and pancreatic bacterial microbiomes in patients with pancreatic cancer and other gastrointestinal diseases. J Oral Microbiol 13:1887680. doi: 10.1080/20002297.2021.1887680 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Farber JM, Peterkin PI. 1991. Listeria monocytogenes, a food-borne pathogen. Microbiol Rev 55:476–511. doi: 10.1128/mr.55.3.476-511.1991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Pourpre R, Lakisic G, Desgranges E, Cossart P, Pagliuso A, Bierne H. 2022. A bacterial virulence factor interacts with the splicing factor RBM5 and stimulates formation of nuclear RBM5 granules. Sci Rep 12:21961. doi: 10.1038/s41598-022-26037-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Bierne H, Tham TN, Batsche E, Dumay A, Leguillou M, Kernéis-Golsteyn S, Regnault B, Seeler JS, Muchardt C, Feunteun J, Cossart P. 2009. Human BAHD1 promotes heterochromatic gene silencing. Proc Natl Acad Sci USA 106:13826–13831. doi: 10.1073/pnas.0901259106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Brena RM, Morrison C, Liyanarachchi S, Jarjoura D, Davuluri RV, Otterson GA, Reisman D, Glaros S, Rush LJ, Plass C. 2007. Aberrant DNA methylation of OLIG1, a novel prognostic factor in non-small cell lung cancer. PLoS Med 4:e108. doi: 10.1371/journal.pmed.0040108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. García E, Marcos-Gutiérrez C, del Mar Lorente M, Moreno JC, Vidal M. 1999. RYBP, a new repressor protein that interacts with components of the mammalian polycomb complex, and with the transcription factor YY1. EMBO J 18:3404–3418. doi: 10.1093/emboj/18.12.3404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Chen D, Zhang J, Li M, Rayburn ER, Wang H, Zhang R. 2009. RYBP stabilizes p53 by modulating MDM2. EMBO Rep 10:166–172. doi: 10.1038/embor.2008.231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Spector DL, Lamond AI. 2011. Nuclear speckles. Cold Spring Harb Perspect Biol 3:a000646. doi: 10.1101/cshperspect.a000646 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Xu Y, Su Z, Li J, Wang Q, Meng G, Zhang Y, Yang W, Zhang J, Gao P. 2019. Role of RNA‑binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer (Review). Oncol Lett 17:2013–2019. doi: 10.3892/ol.2018.9818 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ketter PM, Yu JJ, Guentzel MN, May HC, Gupta R, Eppinger M, Klose KE, Seshu J, Chambers JP, Cap AP, Arulanandam BP. 2018. Acinetobacter baumannii gastrointestinal colonization is facilitated by secretory IgA which is reductively dissociated by bacterial thioredoxin A. mBio 9:e01298-18. doi: 10.1128/mBio.01298-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Moon DC, Gurung M, Lee JH, Lee YS, Choi CW, Kim SI, Lee JC. 2012. Screening of nuclear targeting proteins in Acinetobacter baumannii based on nuclear localization signals. Res Microbiol 163:279–285. doi: 10.1016/j.resmic.2012.02.001 [DOI] [PubMed] [Google Scholar]
- 87. Pećina-Slaus N. 2003. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int 3:17. doi: 10.1186/1475-2867-3-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Na TY, Schecterson L, Mendonsa AM, Gumbiner BM. 2020. The functional activity of E-cadherin controls tumor cell metastasis at multiple steps. Proc Natl Acad Sci USA 117:5931–5937. doi: 10.1073/pnas.1918167117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Chen XH, Wang A, Chu AN, Gong YH, Yuan Y. 2019. Mucosa-associated microbiota in gastric cancer tissues compared with non-cancer tissues. Front Microbiol 10:1261. doi: 10.3389/fmicb.2019.01261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Chakinala RC, Khatri AM. 2023. Gastrointestinal tuberculosis. StatPearls, Treasure Island (FL). [Google Scholar]
- 91. Chow SH, Deo P, Naderer T. 2016. Macrophage cell death in microbial infections. Cell Microbiol 18:466–474. doi: 10.1111/cmi.12573 [DOI] [PubMed] [Google Scholar]
- 92. Lee KK, Workman JL. 2007. Histone acetyltransferase complexes: one size doesn’t fit all. Nat Rev Mol Cell Biol 8:284–295. doi: 10.1038/nrm2145 [DOI] [PubMed] [Google Scholar]
- 93. Barnes AMT, Dale JL, Chen Y, Manias DA, Greenwood Quaintance KE, Karau MK, Kashyap PC, Patel R, Wells CL, Dunny GM. 2017. Enterococcus faecalis readily colonizes the entire gastrointestinal tract and forms biofilms in a germ-free mouse model. Virulence 8:282–296. doi: 10.1080/21505594.2016.1208890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Khan Z, Siddiqui N, Saif MW. 2018. Enterococcus faecalis infective endocarditis and colorectal carcinoma: case of new association gaining ground. Gastroenterology Res 11:238–240. doi: 10.14740/gr996w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Dziubańska-Kusibab PJ, Berger H, Battistini F, Bouwman BAM, Iftekhar A, Katainen R, Cajuso T, Crosetto N, Orozco M, Aaltonen LA, Meyer TF. 2020. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat Med 26:1063–1069. doi: 10.1038/s41591-020-0908-2 [DOI] [PubMed] [Google Scholar]

