Bacterial infection is a major threat to human health, with infections resulting in considerable mortality, urging the need for a more profound understanding of bacteria–host interactions. During infection of cells, host cytoskeletal networks constantly interact with bacteria and are integral to their uptake. Vimentin, an intermediate filament protein, is one such cytoskeletal component that interacts with bacteria during infection. Although vimentin is predominantly present in the cytoplasm, it also appears in a secreted form or at the surface of multiple cell types, including epithelial cells, endothelial cells, macrophages and fibroblasts. As a cytoplasmic protein, vimentin participates in bacterial transportation and the consequential immune-inflammatory responses. When expressed on the cell surface, vimentin can be both pro- and anti-bacterial, favoring bacterial invasion in some contexts, but also limiting bacterial survival in others. Vimentin is also secreted and located extracellularly, where it is primarily involved in bacterial-induced inflammation regulation. Reciprocally, bacteria can also manipulate the fate of vimentin in host cells. Given that vimentin is not only involved in bacterial infection, but also the associated life-threatening inflammation, the use of vimentin-targeted drugs might offer a synergistic advantage. In this Review, we recapitulate the abundant evidence on vimentin and its dynamic changes in bacterial infection and speculate on its potential as an anti-bacterial therapeutic target.

During infection, the interaction between bacteria and host determines the ability of the bacteria to survive and spread (Sukumaran et al., 2021; Yang et al., 2015). Upon invading, bacteria hijack host cells as an environment for survival and transmission, and host cell immune signals are activated for antimicrobial responses (Chisholm et al., 2006; Hooper et al., 2012; Pinaud et al., 2018).

Bacteria–host interactions during invasion include utilization of the host cytoskeleton, of which intermediate filaments (IFs) are an essential component (Mak and Brüggemann, 2016). Compared with other cytoskeletal components, such as actin and microtubules, IFs show a unique combination of flexibility, extensibility and toughness that afford mechanical resilience to the cell (Hu et al., 2019; Patteson et al., 2020a; van Bodegraven and Etienne-Manneville, 2021). These mechanical characteristics protect the cell against damage in various conditions, such as nuclear damage during cell migration (Patteson et al., 2019), shear stress (Flitney et al., 2009; Helmke et al., 2000) and importantly, pathogen infections (Geisler and Leube, 2016).

Vimentin, a type III IF protein, is mainly expressed in mesenchymal cells derived from mesoderm, such as fibroblasts, endothelial cells, neutrophils and macrophages, which interact widely with pathogens (Fuchs and Weber, 1994; Kidd et al., 2014; Mor-Vaknin et al., 2003). Vimentin IFs are formed through polymerization of a single protein, unlike other IFs, such as keratin IFs or neurofilaments, which are copolymers of multiple distinct but related gene products (Herrmann and Aebi, 2016). Vimentin monomers consists of a non-helical amino (N)-terminal head, a conserved central α-helical rod and a carboxy (C)-terminal tail domain. During polymerization, the rod domains first promote the parallel association of monomers into dimers, then dimers bind in an antiparallel manner to form soluble apolar tetramers, considered the structural units for vimentin polymerization. Further lateral assembly of eight tetramers to form unit length filaments (ULFs) is followed by longitudinal annealing and radial compaction to generate insoluble mature vimentin filaments (Herrmann and Aebi, 2016; Zhang et al., 2021).

In recent decades, vimentin has been shown to play both mechanical and non-mechanical roles in the cell through its dynamic changes regulated by post-translational modifications and intracellular proteases (Lowery et al., 2015; Patteson et al., 2020b). Its mechanical functions mainly involve the maintenance of cell morphology, cell adhesion and migration, cell elasticity and subcellular localization of organelles. Meanwhile, its non-mechanical functions are becoming more appreciated, including signaling roles in wound healing and lipogenesis, marking circulating tumor cells and mediating the infection of pathogens (Ivaska et al., 2007; Liu et al., 2019; Mitra et al., 2015; Patteson et al., 2020b; Snider and Omary, 2014; van Engeland et al., 2019). In addition to its function as a cytosolic protein, studies have also revealed the existence of cell surface vimentin (CSV) and secreted vimentin, which are involved in cell activation and inflammatory responses (Adolf et al., 2019; Chi et al., 2012; Ghosh et al., 2018; Lam et al., 2018; Mor-Vaknin et al., 2003).

This Review summarizes the diverse interactions between vimentin and bacteria, highlighting the functional and dynamic changes vimentin undergoes during bacterial infection. We discuss how vimentin participates in the bacterial infection life cycle, focusing mainly on entry and trafficking, as well as the host defense response to invoke inflammation and pathogen clearance. Furthermore, we illustrate how bacterial infection changes the fate of vimentin to further manipulate host cellular processes, including epithelial–mesenchymal transition (EMT), cell differentiation and abnormal inflammatory responses.

As an important component of the cytoskeleton, vimentin forms a unique filamentous network in the cytoplasm. During bacterial infection, cytoplasmic vimentin regulates bacterial transport and mediates inflammatory signal transduction (Gasse et al., 2009). We summarize the interactions between cytoplasmic vimentin and bacteria and the involved virulence factors (Table 1).

Table 1.

Summary of the interactions between bacteria and the distinct forms of vimentin

Summary of the interactions between bacteria and the distinct forms of vimentin
Summary of the interactions between bacteria and the distinct forms of vimentin

Cytoplasmic vimentin in bacterial transportation

During invasion, bacteria typically hijack the host cytoskeleton for further trafficking. To escape detection from immune signals and/or degradation by lysosomes, many bacteria are transported in a form of vacuole called bacteria-containing vacuoles (BCVs) (Creasey and Isberg, 2014), and cytoplasmic vimentin was found mainly to regulate bacterial transportation and replication via BCVs. For example, Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative intracellular pathogen that causes gastroenteritis in susceptible humans and fatal typhoid-like affliction in mice (Tsolis et al., 2000). After invading the host cell, these bacteria enter a vacuole, named the Salmonella-containing vacuole (SCV). The bacteria begin to replicate when the SCV reaches the juxtanuclear area. It has been shown that vimentin networks aggregate around SCVs from 7 h post-infection onwards (Guignot and Servin, 2008) (Fig. 1). Importantly, when vimentin is knocked down, SCVs become scattered throughout the cytoplasm, indicating that vimentin is critical in maintaining SCVs in the juxtanuclear area (Guignot and Servin, 2008). These results are consistent with the common role of vimentin in controlling the structure and localization of subcellular organelles (Toivola et al., 2005). The S. Typhimurium effector protein SptP also interacts with cytoplasmic vimentin, inducing recruitment of vimentin to the Salmonella-induced membrane ruffles (Murli et al., 2001). Vimentin is also key in Chlamydia trachomatis replication within BCVs. C. trachomatis vacuoles are stabilized by a scaffolding network made of F-actin and vimentin filaments, which encapsulate the vacuoles (Kumar and Valdivia, 2008) (Fig. 1). The protease Chlamydia protease-like activity factor (CPAF), secreted by C. trachomatis, targets and cleaves the head domain of vimentin and alters its properties, thus regulating the toughness and flexibility of vacuoles (Kumar and Valdivia, 2008; Snider et al., 2018; Tarbet et al., 2018). Another bacterium that utilizes vacuoles for replication is Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis disease, one of the most common tick-borne diseases (Stuen et al., 2013). This unusual obligate intracellular pathogen selectively adheres to polymorphonuclear leukocytes, the most abundant innate immune cells in the circulation and one of the most important lines of defense in innate immunity (Nicu and Loos, 2016; Yago et al., 2003). In infected cells, A. phagocytophilum resides and duplicates within A. phagocytophilum-occupied vacuoles (ApVs). Studies have shown that both vimentin and keratin IF proteins aggregate to surround ApVs. In addition, polySUMOylation, the attachment of small ubiquitin-like modifier (SUMO) chains (Yang and Zhang, 2014), increases the solubility of vimentin, which contributes to its rearrangement around ApVs (Fig. 1). Consistent with this, more vimentin exists in an insoluble state in A. phagocytophilum-infected cells than in uninfected cells. The bacterial load is significantly reduced when cytoplasmic vimentin is inhibited by Withaferin A (WFA), which binds to soluble vimentin and causes its aggregation (Bargagna-Mohan et al., 2007). Therefore, A. phagocytophilum modulate vimentin to construct their vacuolar niche and promote optimal survival (Truchan et al., 2016). In addition, vimentin surrounding ApVs interacts with A. phagocytotopulum toxin A protein (AptA) to activate the mammalian ERK1 and ERK2 (also known as MAPK3 and MAPK1, respectively) mitogen-activated protein kinase (MAPK), which is essential for A. phagocytophilum survival (Sukumaran et al., 2011; Xiong et al., 2009; Xiong and Rikihisa, 2011). Thus, cytoplasmic vimentin is involved in both the mechanical regulation of BCVs, as well as cell signaling events during bacterial infection. For a summary of these interactions, see Table 1.

Fig. 1.

Schematic illustration of how vimentin mediates bacterial entry and trafficking. CSV interacts with bacterial effectors to mediate bacterial entry. Highlighted are factors that mediate bacterial entry. Factors that mediates entry into brain BMECs are IbeA (E. coli K1 entry), BspC (GBS entry) and InIF (L. monocytogenes entry). CSV also interacts with MBP-1 during MAH entry into human respiratory epithelial cells, and the primary effector IpaC acts to promote the stable docking between S. flexneri and mouse embryonic fibroblasts. Cytoplasmic vimentin mediates bacterial trafficking. The S. Typhimurium effector protein SptP recruits cytoplasmic vimentin to Salmonella-induced ruffles, and cytoplasmic vimentin mediates the trafficking of SCVs to the juxtanuclear area. C. trachomatis-containing vacuoles are stabilized by vimentin scaffolds; here, cytoplasmic vimentin rearranges around the ApV, and the A. phagocytotopulum effector AptA interacts with vimentin to promote optimal bacterial survival in the vacuole niche.

Fig. 1.

Schematic illustration of how vimentin mediates bacterial entry and trafficking. CSV interacts with bacterial effectors to mediate bacterial entry. Highlighted are factors that mediate bacterial entry. Factors that mediates entry into brain BMECs are IbeA (E. coli K1 entry), BspC (GBS entry) and InIF (L. monocytogenes entry). CSV also interacts with MBP-1 during MAH entry into human respiratory epithelial cells, and the primary effector IpaC acts to promote the stable docking between S. flexneri and mouse embryonic fibroblasts. Cytoplasmic vimentin mediates bacterial trafficking. The S. Typhimurium effector protein SptP recruits cytoplasmic vimentin to Salmonella-induced ruffles, and cytoplasmic vimentin mediates the trafficking of SCVs to the juxtanuclear area. C. trachomatis-containing vacuoles are stabilized by vimentin scaffolds; here, cytoplasmic vimentin rearranges around the ApV, and the A. phagocytotopulum effector AptA interacts with vimentin to promote optimal bacterial survival in the vacuole niche.

Role of cytoplasmic vimentin in inflammation and immune responses upon bacterial infection

Vimentin displays a wide range of non-mechanical functions in the process of apoptosis and inflammation (Su et al., 2019a; Su et al., 2019b). In inflammatory and autoimmune diseases (Mortensen et al., 2015), vimentin can serve as a scaffold to coordinate immune defenses. This includes the localization of a number of pattern recognition receptors (PRRs), which mediate initial sensing of infection and the subsequent activation of the inflammatory response (Dos Santos et al., 2015; Mizushima and Levine, 2020; Thiagarajan et al., 2013) (Fig. 2), as well as autophagy, an important line of defense against pathogens in eukaryotes (Kroemer et al., 2010; Mizushima and Levine, 2020). Vimentin regulates autophagy by altering the re-localization of autophagosomes from the cytoplasm to the juxtanuclear region, given that treatment with the vimentin inhibitor WFA inhibits the fusion of autophagosomes and lysosomes, thus preventing their maturation autolysosomes (Biskou et al., 2019).

Fig. 2.

Schematic illustration of vimentin functions during bacteria-induced inflammatory responses. CSV is crucial for the invasion of P. acnes and AIEC by inducing an NF-κB- and NOD2-mediated inflammatory response in macrophages (blue arrows). Cytoplasmic vimentin filaments serve as scaffolds for NLRP3 inflammasomes and NOD2. Upon LPS exposure, NLRP3 inflammasomes interact with and are activated by cytoplasmic vimentin, with subsequent maturation of IL-1β (green arrows). Additionally, ROS elevation promotes vimentin expression, as well as activation of NF-κB signaling. However, infection with Mtb results in a reduction in the level of vimentin and ROS via ESAT-6; this subsequently suppresses the activation of NLPR3 and NF-κB, accompanied by lower levels of pro-inflammatory cytokines (yellow arrows). Finally, secretion of vimentin is triggered by the pro-inflammatory cytokine TNF and blocked by the anti-inflammatory cytokine IL-10 (purple arrows).

Fig. 2.

Schematic illustration of vimentin functions during bacteria-induced inflammatory responses. CSV is crucial for the invasion of P. acnes and AIEC by inducing an NF-κB- and NOD2-mediated inflammatory response in macrophages (blue arrows). Cytoplasmic vimentin filaments serve as scaffolds for NLRP3 inflammasomes and NOD2. Upon LPS exposure, NLRP3 inflammasomes interact with and are activated by cytoplasmic vimentin, with subsequent maturation of IL-1β (green arrows). Additionally, ROS elevation promotes vimentin expression, as well as activation of NF-κB signaling. However, infection with Mtb results in a reduction in the level of vimentin and ROS via ESAT-6; this subsequently suppresses the activation of NLPR3 and NF-κB, accompanied by lower levels of pro-inflammatory cytokines (yellow arrows). Finally, secretion of vimentin is triggered by the pro-inflammatory cytokine TNF and blocked by the anti-inflammatory cytokine IL-10 (purple arrows).

Acute lung injury (ALI) is associated with activation of the NOD-like receptor protein 3 (NLRP3) inflammasome and subsequent maturation of interleukin (IL)-1β, which leads to excessive inflammation (Gasse et al., 2009). It has been reported that vimentin colocalizes with NLRP3 and active caspase-1 during the onset of ALI (Gasse et al., 2009). In addition, the absence of vimentin suppresses the pathophysiological events of ALI in mice, including IL-1β expression, endothelial and alveolar epithelial barrier permeability and fibrosis, which are induced by lipopolysaccharides (LPS), bleomycin and asbestos, respectively (Dos Santos et al., 2015; Zhou et al., 2020). In contrast, vimentin knockdown in lung fibroblasts results in increased apoptosis and inflammatory cytokines, including tumor necrosis factor (TNF; also known as TNF-α), IL-1β, IL-6 and IL-10, when challenged with LPS (Pan et al., 2021). Whether this result differs due to cell-specific effects is worth further inquiry.

When challenged by LPS, caspase-3 is significantly upregulated in vimentin-deficient Jurkat T cells and reciprocally downregulated in vimentin-overexpressing cells (Su et al., 2019a). Levels of the cytokines IL-2, IL-10 and interferon γ (IFN-γ) are significantly lower in vimentin-deficient cells, whereas they are unchanged in vimentin-overexpressing cells (Su et al., 2019a), perhaps because the endogenous vimentin levels are already enough to help induce high levels of these cytokines when challenged by LPS. Moreover, vimentin depletion significantly enhances the metabolic and suppressive activity of regulatory T cells (Tregs) (McDonald-Hyman et al., 2018), although this was in the context of graft-versus-host disease, and not bacterial infection.

In addition to macrophage activation, differentiation and phagocytosis, vimentin is also involved in reactive oxygen species (ROS) production (Beneš et al., 2006; Mor-Vaknin et al., 2003). Mycobacterium tuberculosis (Mtb) is a highly adaptive pathogen that evolved with humans (Brites and Gagneux, 2015). Treating Mtb-infected macrophages with hydrogen peroxide (H2O2), a type of ROS, increases vimentin expression (Mahesh et al., 2016). In contrast, vimentin expression decreases in Mtb-infected macrophages treated with N-acetyl L-cysteine (NAC), a scavenger of ROS, suggesting vimentin expression is ROS dependent (Mahesh et al., 2016). Moreover, ectopic expression of the Mtb protein early secreted antigenic target of 6 kDa (ESAT-6) decreases both the level of ROS and the expression of vimentin in macrophages, which implies that Mtb-mediated downregulation of vimentin is, at least in part, due to the downregulation of ROS by the pathogen (Mahesh et al., 2016; Mishra et al., 2010). Interestingly, although ESAT-6 reduces vimentin expression, both ESAT-6 and vimentin are involved in the assembly of the NLRP3 inflammasome (Dos Santos et al., 2015), which is worthy of more investigation. In addition, ROS are involved in nuclear factor (NF)-κB activation and pro-inflammatory responses induced by LPS (Gloire et al., 2006). Interestingly, the incubation of macrophages with anti-vimentin antibodies increases the production of ROS (Mahesh et al., 2016). Additionally, vimentin-deficient phagocytes or vimentin-knockout mice challenged with Escherichia coli in the abdominal cavity produce more ROS and nitric oxide (NO) to kill bacteria (Mor-Vaknin et al., 2013). Together, these findings demonstrate that vimentin is a key metabolic and functional regulator of inflammatory responses, particularly involving ROS (Fig. 2), and provide evidence that regulation of vimentin might be used to treat bacterial infections.

Although originally identified as a cytoplasmic protein (Steinert and Liem, 1990), vimentin has since been shown to also localize to the cell surface (Dupont et al., 2004; Segawa et al., 2005). Previous studies indicate that CSV mediates the landing of bacteria at specific membrane sites, where it acts as a receptor and/or coreceptor. Moreover, CSV has been found to participate in limiting bacterial survival (Stevens et al., 2013). In this section, we describe how CSV can both enhance bacterial infection and limit bacterial survival.

Involvement of CSV in bacterial entry

Meningitis, the inflammation of the meninges with high morbidity and mortality rates (Putz et al., 2013), is the most common serious infection in newborns (van der Flier, 2021). The pathogenic bacteria of meningitis include E. coli K1, group B Streptococcus (GBS) and Listeria monocytogenes, among others (Kim et al., 2009). The blood–brain barrier (BBB) is an important structure protecting against the invasion of pathogens into the brain and preventing meningitis (Huang et al., 2000; Kim, 2003). The BBB is formed by brain microvascular endothelial cells (BMECs), which have high endogenous vimentin expression (Zou et al., 2006). It has been reported that the interaction between IbeA, an E coli K1 effector, with the head domain of CSV is critical for E coli K1 invasion (Chi et al., 2010; Zou et al., 2006). In addition, IbeA and IbeA+ E coli K1 are able to induce clustering of vimentin at the surface of BMECs, which is required for the invasion process (Chi et al., 2010). GBS infection, the leading cause of meningitis, continues to increase in incidence and affects a wider age group with more diverse clinical manifestations, including skin, soft tissue, heart valve and urinary tract infections (Raabe and Shane, 2019). CSV interacts with BspC, the GBS surface factor, via its tail domain to promote bacterial adhesion to BMECs (Beninati et al., 2019; Deng et al., 2019). L. monocytogenes also utilize CSV as an invasion receptor to interact with the virulence factor InIF and promote bacterial invasion (Ghosh et al., 2018). In a mouse model deficient in vimentin, L. monocytogenes colonization in the brain is severely compromised (Ghosh et al., 2018). Another study found that extracellular matrix stiffness can regulate the susceptibility of mammalian host cells to L. monocytogenes infection and that this regulation depends on the level of CSV, which acted as host cell surface receptors (Bastounis et al., 2018).

In addition to meningitis-related pathogens, CSV is also critical for the invasion of other bacteria. Propionibacterium acnes was initially identified as being an important member of the skin microbiome and associated with acne vulgaris (Byrd et al., 2018). However, there is evidence suggesting it is related to other chronic diseases, such as prostate inflammation, including upregulation of inflammatory cytokines and chemokines (Perry and Lambert, 2011). P. acnes can effectively penetrate prostate cells expressing CSV. Antibody-mediated neutralization of CSV unveils the role of vimentin as a cell surface mediator for invasion of P. acnes (Mak et al., 2012).

CSV is also important for invasion of the Mycobacterium avium subspecies hominissuis (MAH), which causes respiratory diseases in immunocompromised patients, such as cystic fibrosis and chronic respiratory diseases (Daley, 2017). During initial airway colonization, MAH forms microaggregates consisting of three to 20 bacterial species on human respiratory epithelial cells via microaggregate-binding protein 1 (MBP-1; also known as general stress protein CsbD, encoded by MAV_3013), which leads to effective invasion towards mucous membranes (Babrak et al., 2015). Interestingly, vimentin interacts with MBP-1, and anti-vimentin antibody V9 blocks the binding of microaggregates to host cells (Babrak et al., 2015). Shigella flexneri employs the type III secretion system (T3SS) to inject effector proteins into target host cells, enabling bacterial infection (Puhar and Sansonetti, 2014; Ramos-Morales, 2012; Zhou and Galán, 2001). Invasion plasmid antigen C (IpaC), secreted via the T3SS, has been identified as the primary effector protein for epithelial cell invasion (Duncan-Lowey et al., 2020). CSV has been shown to interact with IpaC, which is required for the stable docking of S. flexneri to host cells and further triggers effector secretion (Russo et al., 2016). Together, the above studies conclude that bacteria invade host cells through interactions between key virulence factors and specific domains of CSV. Identifying the exact interaction sites requires further exploration before their interruption might be used as a potential target for therapy.

Role of CSV in bacterial clearance by mediating inflammatory responses

The initial sensing of a pathogenic infection is mediated by innate PRRs, which recognize and bind to pathogen-associated molecular patterns (PAMPs) to trigger the activation of inflammatory and antimicrobial signals (Takeuchi and Akira, 2010). Nucleotide binding oligomerization domain-containing protein 2 (NOD2), as an intracellular PRR, binds to the ligand muramyl dipeptide (MDP) to induce pathogen transmission and clearance (Caruso et al., 2014). NOD2 mutations remain the strongest single genetic determinant of Crohn's disease (CD). Three disease-associated polymorphisms of NOD2, including R702W, G908R and L1007fs, all alter the leucine-rich repeat domain (LRR) of NOD2 (Packwood et al., 2010). Interestingly, NOD2 interacts with CSV through its LRR domain, and this interaction affects NOD2 localization on the cell membrane (Stevens et al., 2013). Moreover, NOD2 and CSV regulate the invasiveness and survival of adherent-invasive E. coli (AIEC) (Stevens et al., 2013). The vimentin inhibitor WFA effectively impairs the ability of NOD2 to limit bacterial survival by inhibiting NOD2-dependent NF-κB activation and autophagy (Stevens et al., 2013). These studies suggest that vimentin favors limiting bacterial survival by cooperating with NOD2 to mediate the inflammation responses. Moreover, vimentin expressed on the surface of apoptotic neutrophils can be used as an ‘eat me’ signal that is recognized by phagocytes (Moisan and Girard, 2006). It is worth mentioning that, although vimentin limits bacterial survival using the above mechanisms, depletion of vimentin or inhibition by WFA can also reduce bacterial invasion (Henderson and Stevens, 2012; Henderson et al., 2012; Stevens et al., 2013). Several studies support a pro-infection role of vimentin, but only few reports describe the role of vimentin in restricting bacterial infection. Therefore, the precise and complex antagonistic relationship between bacteria and cellular vimentin deserves further investigation.

Secreted vimentin and immune cells

Macrophages can secrete vimentin and this process is regulated by inflammatory factors (Mor-Vaknin et al., 2003); the anti-inflammatory cytokine IL-10 blocks vimentin secretion, whereas the pro-inflammatory cytokine TNF triggers vimentin secretion (Mor-Vaknin et al., 2003). During bacterial infection, secreted vimentin promotes innate inflammatory responses by increasing the bactericidal capacity of macrophages and the level of active oxidative metabolite production (Mor-Vaknin et al., 2003). The Clostridium botulinum C3 exoenzyme (C3bot) is a bacterial ADP-ribosomal transferase that inhibits Rho family small GTPases. It has been used as a pharmacological tool to investigate cellular Rho function and to repair neuronal injury (Loske et al., 2012). Astrocytes have been shown to secrete exosomes containing vimentin, which facilitates C3bot interactions with neurons and neuronal recovery after injury (Adolf et al., 2019). Another study has shown that secreted vimentin promotes LPS-induced activation of human dendritic cells by regulating the secretion of IL-6, IL-12 and IL-10 (Fig. 2). Among them, the anti-inflammatory cytokine IL-10 prevents the differentiation of T cells to Th1 cells, thereby reducing the development of tissue damage caused by autoimmunity (Yu et al., 2018). The interactions between secreted vimentin, bacteria and the virulence factors are summarized in Table 1.

Neutrophils, the most abundant type of leukocyte (white blood cell) in the body, protect the body from harmful microbial infections, especially those caused by bacterial and fungal pathogens (van Rees et al., 2016). Neutrophils tend to adhere to endothelial cells via P-selectin, an early step in acute inflammatory responses (Liou et al., 2013). However, excessive activation of leukocytes causes autoimmune diseases, including ALI (Lam et al., 2018). Platelets can repair damage and maintain vascular integrity through adhesion, that is, platelet–platelet interaction (Berger et al., 1998). Recombinant vimentin has been shown to decrease the adhesion of neutrophils to the platelet receptor P-selectin, thereby reducing ALI (Lam et al., 2018). It has been speculated that secreted vimentin has the same ability. Whether secreted vimentin behaves similarly to recombinant vimentin is unknown. Several studies have reported the presence of circulating vimentin in peripheral blood (Bay-Jensen et al., 2013; Li et al., 2017). Therefore, whether naturally occurring circulating vimentin also participates in immune function through P-selectin, and thus regulation of leukocyte–platelet interactions, deserves further investigation.

Post-translational regulation of secreted vimentin by citrullination

Citrullination, the conversion of the amino acid arginine to citrulline, is catalyzed by peptidyl arginine deiminases (PADs) (Vossenaar et al., 2004). Neutrophil extracellular traps (NETs), released via a form of cell death called NETosis, consist of a chromatin meshwork decorated with antimicrobial peptides typically present in neutrophil granules (Brinkmann et al., 2004). NETs bind microorganisms, degrade virulence factors and kill bacteria (Brinkmann et al., 2004; Hirschfeld et al., 2017; Mutua and Gershwin, 2021). During NETosis, citrullinated vimentin is externalized into NETs (Khandpur et al., 2013). Interestingly, rheumatoid arthritis (RA) is characterized by the breakdown of self-tolerance to citrulline antigens, producing autoantibodies to citrullinated protein antigens (ACPAs) (Corsiero et al., 2016; Klareskog et al., 2014). Citrullinated vimentin is one of the candidate antigens (Corsiero et al., 2016; Luime et al., 2010; Van Steendam et al., 2011). Notably, RA serum and immunoglobulin fractions significantly enhance NETosis in RA patients with high ACPA levels (Khandpur et al., 2013). Neutrophils externalize citrullinated autoantigens associated with RA pathogenesis, and anti-citrullinated vimentin antibodies can effectively induce NET formation (Khandpur et al., 2013). In addition, NETs significantly enhance the inflammatory response in RA and osteoarthritis synovial fibroblasts, including the induction of IL-6, IL-8, chemokines and adhesion molecules (Khandpur et al., 2013). These observations suggest that enhanced NETosis, with externalized citrullinated vimentin, promotes abnormal adaptive and innate immune responses in joints and the periphery during RA pathogenesis and perpetuates the pathogenic mechanisms of the disease (Khandpur et al., 2013).

In addition to participating in bacterial infection, vimentin in host cells can also be adversely affected by various bacteria (Table 1). Vimentin is often used as a marker of mesenchymal-derived cells, or cells undergoing EMT during normal development and metastatic progression (Satelli and Li, 2011). S. Typhimurium specifically targets microfold cells (M cells), which in contrast to other intestinal cells, express vimentin (Kraehenbuhl and Neutra, 2000; Tahoun et al., 2011). The T3SS effector protein SopB of S. Typhimurium increases the expression levels of host cell vimentin, which induces the transformation of follicular-associated epithelium (FAE) into M cells (Tahoun et al., 2012). This promotes bacterial infection by activating the Wnt/β-catenin and NF-κB signaling pathways (Tahoun et al., 2012). This is also exemplified by H. pylori, a bacterium found on epithelial cells in the gastric mucosa (Huang et al., 2017). Studies have shown that vimentin expression increases in H. pylori-infected cells, inducing EMT-like phenotypic changes in gastric cancer, which contribute to gastric carcinogenesis (Fig. 3) (Bessède et al., 2014; Lamouille et al., 2014; Marques et al., 2018; Wroblewski et al., 2015; Yu et al., 2014). A recent study has shown that intracellular bacteria in breast tumor cells promote tumor metastasis by reducing the density of actin stress fibers (Fu et al., 2022). Notably, vimentin is a marker of EMT, and numerous studies have shown that its expression increases during bacterial infection-induced carcinogenesis as well as increasing hematogenous metastasis of tumors (Abd-El-Raouf et al., 2020; Chen et al., 2020; Choi et al., 2015; Hofman and Vouret-Craviari, 2012). Whether vimentin is involved in intracellular bacteria-induced tumor metastasis deserves further attention.

Fig. 3.

Schematic illustration of bacteria-mediated regulation of vimentin after infection. Overview of bacterial factors that regulate vimentin rearrangements. The E. coli K1 effector IbeA induces vimentin clustering in BMECs, and the MAH effector MBP-1 induces vimentin aggregation in human respiratory epithelial cells. Other bacteria induce vimentin disassembly; the P. aeruginosa effector ExoA destroys the filamentous structure of vimentin and its effector ExoS disassembles vimentin by catalyzing ADP ribosylation. Furthermore, the protease CPAF, secreted by C. trachomatis, cleaves the head domain of vimentin and alters its cytoskeletal properties. Finally, SpyA is an ADP ribotransferase secreted by S. pyogenes that modifies the arginine head domain of vimentin, causing its tetramer subunit to collapse. Some bacteria promote vimentin expression to induce host cell EMT. For example, the effector protein SopB of S. Typhimurium induces a transition from follicular-associated epithelium into M cells, which are marked by vimentin, whereas H. pylori CagA induces EMT-associated changes in gastric cancer, including elevated vimentin expression, which results in increased hematogenous metastasis of tumors.

Fig. 3.

Schematic illustration of bacteria-mediated regulation of vimentin after infection. Overview of bacterial factors that regulate vimentin rearrangements. The E. coli K1 effector IbeA induces vimentin clustering in BMECs, and the MAH effector MBP-1 induces vimentin aggregation in human respiratory epithelial cells. Other bacteria induce vimentin disassembly; the P. aeruginosa effector ExoA destroys the filamentous structure of vimentin and its effector ExoS disassembles vimentin by catalyzing ADP ribosylation. Furthermore, the protease CPAF, secreted by C. trachomatis, cleaves the head domain of vimentin and alters its cytoskeletal properties. Finally, SpyA is an ADP ribotransferase secreted by S. pyogenes that modifies the arginine head domain of vimentin, causing its tetramer subunit to collapse. Some bacteria promote vimentin expression to induce host cell EMT. For example, the effector protein SopB of S. Typhimurium induces a transition from follicular-associated epithelium into M cells, which are marked by vimentin, whereas H. pylori CagA induces EMT-associated changes in gastric cancer, including elevated vimentin expression, which results in increased hematogenous metastasis of tumors.

Several bacterial factors are reported to interfere with vimentin to regulate its assembly and disassembly through post-translational modifications (Icenogle et al., 2012). SpyA is a streptococcal ADP ribosyltransferase that modifies the arginine head domain of vimentin (including arginine residues 44, 49 and 63), to cause the collapse of the tetramer subunit of vimentin (Fig. 3) (Coye and Collins, 2004; Icenogle et al., 2012; Snider and Omary, 2014). SpyA also triggers caspase-1-dependent inflammatory response and reduces filamentous vimentin in macrophages (Icenogle et al., 2012; Lin et al., 2015). In addition, the effector protein Exotoxin S (ExoS) of Pseudomonas aeruginosa can catalyze ADP-ribosylation and disassemble vimentin (Coburn et al., 1989; Kroken et al., 2022). Exotoxin A (ExoA) of P. aeruginosa has been shown to disrupt the filamentous structure of vimentin, while causing no discernible effect on microtubules or microfilaments (Sharpe et al., 1980).

As a dynamic structure, vimentin can be regulated by cell cycle-dependent endogenous phosphorylation, which usually occurs on its N-terminal head domain, facilitating filament disassembly. This process is mediated by protein kinases A (PKA) and C (PKC), calmodulin (CaM)-kinase II, p21-activated kinase (PAK) and Cdc2 kinase (Ku et al., 2002; Snider and Omary, 2014). S. Typhimurium effector SptP is a tyrosine phosphatase that can de-phosphorylate vimentin to make it more flexible (Murli et al., 2001). Both SptP and vimentin are recruited to membrane ruffles formed by the actin cytoskeleton (Fig. 3) (Fu and Galán, 1998; Kraxner et al., 2021; Murli et al., 2001). HopQ is an effector protein of the plant bacterial pathogen Pseudomonas syringae pv. tomato (Pst). Overexpression of HopQ enhances its interaction with vimentin, allowing vimentin to be degraded by p62-dependent selective autophagy. Attenuation of vimentin expression by HopQ inhibits the motility and metastasis of melanoma in vivo (Park et al., 2020). Together, these findings suggest that bacteria-induced remodeling of vimentin and its related signaling pathways influence the fate of host defense processes as well as of tumor progression.

Vimentin, with its different subcellular localizations, exerts a number of roles during bacterial infection. First, cytoplasmic vimentin mainly works in two ways after cell invasion – bacteria utilize vimentin-coated vacuoles for their intracellular transportation, allowing them to escape from immune surveillance and lysosomal phagocytosis (Guignot and Servin, 2008; Kumar and Valdivia, 2008; Truchan et al., 2016). In addition, cytoplasmic vimentin activates inflammasomes and inflammatory cells upon bacterial infection (Dos Santos et al., 2015; McDonald-Hyman et al., 2018; Mor-Vaknin et al., 2013; Rivera, 2019; Su et al., 2019a; Su et al., 2019b). Second, cell-surface-exposed vimentin mediates bacterial invasion by serving as either a receptor, co-receptor or simply as a preferred landing site (Beninati et al., 2019; Chi et al., 2012; Chi et al., 2010; Deng et al., 2019; Ghosh et al., 2018; Zou et al., 2006). Moreover, CSV also participates in the transmission of pathogen invasion signals by acting as a scaffold for PRR, influencing inflammatory response (Henderson et al., 2012; Stevens et al., 2013). Finally, secreted vimentin has several roles in inflammatory responses, such as promoting the bactericidal function of macrophages (Mor-Vaknin et al., 2003), and reducing the tropism and adhesion of neutrophils, thereby reducing ALI (Lam et al., 2018).

When bacteria invade, they inject virulence factors into the host cytoplasm, which changes the fate of vimentin. Firstly, the expression of vimentin is altered, which leads to an EMT-like transition in infected cells (Abd-El-Raouf et al., 2020; Bessède et al., 2014; Fu et al., 2022; Marques et al., 2018; Tahoun et al., 2012; Wroblewski et al., 2015; Yu et al., 2014). Secondly, the dynamic assembly and disassembly of the vimentin network are coupled with bacterial infection (Coye and Collins, 2004; Icenogle et al., 2012; Ku et al., 2002), which is accompanied by increased bacterial susceptibility, cell differentiation and EMT (Krzysiek-Maczka et al., 2018; Lee et al., 2017; Tahoun et al., 2012; Thanaphongdecha et al., 2020).

In general, regardless of the intricate role in inflammation, vimentin favors bacterial binding and entry by directly serving as a receptor or co-receptor, or by interacting with bacterial proteins as a cell surface protein. Vimentin also assists bacterial trafficking and replication by aggregating around BCVs as a cytoplasmic protein (summarized in Table 1). Strikingly, vimentin also tends to favor viral binding and entry, fusion and replication (Gladue et al., 2013; Nédellec et al., 1998; Pagani et al., 2017; Zhang et al., 2015; Zhang et al., 2021), suggesting a potentially conserved feature in both bacterial infection and viral infection. Especially, converging evidence shows that vimentin plays two distinct roles in COVID-19, not only being involved in the viral infection but also in the associated life-threatening lung inflammation (Li et al., 2020). Moreover, we have previously shown that vimentin serves as a structural organizer and an RNA-binding protein regulator to facilitate Zika virus (ZIKV) replication (Zhang et al., 2022), where we observed substantial reorganization of vimentin surrounding and protecting the ZIKV replication factory. Taken together, all these considerations strongly suggest that vimentin represents a valuable therapeutic target not only in bacterial infections but also in infections more broadly (Zhang et al., 2021), and molecules downregulating vimentin or suppressing the aggregation of vimentin to the pathogen-containing vacuole are promising for treatment of broad-spectrum pathogen infection.

In conclusion, although vimentin has been shown to be important in a number of bacterial infections, its regulatory role and exact molecular mechanisms need to be further elucidated. This will undoubtedly improve our knowledge of pathogen–host interactions, which is necessary if vimentin is to be considered as a potential therapeutic target.

Funding

This work was supported by National Key Research and Development Program of China (2022YFC2303502, 2021YFC2300204); National Natural Science Foundation of China (32222022, 92054104, 31970660); CAS-VPST Silk Road Science Fund (GJHZ2021138); Shanghai Municipal Science and Technology Major Project (2019SHZDZX02).

Abd-El-Raouf
,
R.
,
Ouf
,
S. A.
,
Gabr
,
M. M.
,
Zakaria
,
M. M.
,
El-Yasergy
,
K. F.
and
Ali-El-Dein
,
B.
(
2020
).
Escherichia coli foster bladder cancer cell line progression via epithelial mesenchymal transition, stemness and metabolic reprogramming
.
Sci. Rep.
10
,
18024
.
Adolf
,
A.
,
Rohrbeck
,
A.
,
Münster-Wandowski
,
A.
,
Johansson
,
M.
,
Kuhn
,
H. G.
,
Kopp
,
M. A.
,
Brommer
,
B.
,
Schwab
,
J. M.
,
Just
,
I.
,
Ahnert-Hilger
,
G.
et al. 
(
2019
).
Release of astroglial vimentin by extracellular vesicles: Modulation of binding and internalization of C3 transferase in astrocytes and neurons
.
Glia
67
,
703
-
717
.
Babrak
,
L.
,
Danelishvili
,
L.
,
Rose
,
S. J.
,
Kornberg
,
T.
and
Bermudez
,
L. E.
(
2015
).
The Environment of “Mycobacterium avium subsp. hominissuis” microaggregates induces synthesis of small proteins associated with efficient infection of respiratory epithelial cells
.
Infect. Immun.
83
,
625
-
636
.
Bargagna-Mohan
,
P.
,
Hamza
,
A.
,
Kim
,
Y.-E.
,
Khuan Abby Ho
,
Y.
,
Mor-Vaknin
,
N.
,
Wendschlag
,
N.
,
Liu
,
J.
,
Evans
,
R. M.
,
Markovitz
,
D. M.
,
Zhan
,
C.-G.
et al. 
(
2007
).
The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin
.
Chem. Biol.
14
,
623
-
634
.
Bastounis
,
E. E.
,
Yeh
,
Y.-T.
and
Theriot
,
J. A.
(
2018
).
Matrix stiffness modulates infection of endothelial cells by Listeria monocytogenes via expression of cell surface vimentin
.
Mol. Biol. Cell
29
,
1571
-
1589
.
Bay-Jensen
,
A. C.
,
Karsdal
,
M. A.
,
Vassiliadis
,
E.
,
Wichuk
,
S.
,
Marcher-Mikkelsen
,
K.
,
Lories
,
R.
,
Christiansen
,
C.
and
Maksymowych
,
W. P.
(
2013
).
Circulating citrullinated vimentin fragments reflect disease burden in ankylosing spondylitis and have prognostic capacity for radiographic progression
.
Arthritis. Rheum.
65
,
972
-
980
.
Beneš
,
P.
,
Macečková
,
V.
,
Zdráhal
,
Z.
,
Konečná
,
H.
,
Zahradníčková
,
E.
,
Mužík
,
J.
and
Šmarda
,
J.
(
2006
).
Role of vimentin in regulation of monocyte/macrophage differentiation
.
Differentiation
74
,
265
-
276
.
Beninati
,
C.
,
Famà
,
A.
and
Teti
,
G.
(
2019
).
How BspC from Streptococcus agalactiae Interacts with Host Vimentin during Meningitis
.
Trends Microbiol.
27
,
727
-
728
.
Berger
,
G.
,
Hartwell
,
D. W.
and
Wagner
,
D. D.
(
1998
).
P-Selectin and platelet clearance
.
Blood
92
,
4446
-
4452
.
Bessède
,
E.
,
Staedel
,
C.
,
Acuña Amador
,
L. A.
,
Nguyen
,
P. H.
,
Chambonnier
,
L.
,
Hatakeyama
,
M.
,
Belleannée
,
G.
,
Mégraud
,
F.
and
Varon
,
C.
(
2014
).
Helicobacter pylori generates cells with cancer stem cell properties via epithelial-mesenchymal transition-like changes
.
Oncogene
33
,
4123
-
4131
.
Biskou
,
O.
,
Casanova
,
V.
,
Hooper
,
K. M.
,
Kemp
,
S.
,
Wright
,
G. P.
,
Satsangi
,
J.
,
Barlow
,
P. G.
and
Stevens
,
C.
(
2019
).
The type III intermediate filament vimentin regulates organelle distribution and modulates autophagy
.
PLoS ONE
14
,
e0209665
.
Brinkmann
,
V.
,
Reichard
,
U.
,
Goosmann
,
C.
,
Fauler
,
B.
,
Uhlemann
,
Y.
,
Weiss
,
D. S.
,
Weinrauch
,
Y.
and
Zychlinsky
,
A.
(
2004
).
Neutrophil extracellular traps kill bacteria
.
Science (New York, N.Y.)
303
,
1532
-
1535
.
Brites
,
D.
and
Gagneux
,
S.
(
2015
).
Co-evolution of Mycobacterium tuberculosis and Homo sapiens
.
Immunol. Rev.
264
,
6
-
24
.
Byrd
,
A. L.
,
Belkaid
,
Y.
and
Segre
,
J. A.
(
2018
).
The human skin microbiome
.
Nat. Rev. Microbiol.
16
,
143
-
155
.
Caruso
,
R.
,
Warner
,
N.
,
Inohara
,
N.
and
Núñez
,
G.
(
2014
).
NOD1 and NOD2: signaling, host defense, and inflammatory disease
.
Immunity
41
,
898
-
908
.
Chen
,
J.
,
Gong
,
G.
,
Zheng
,
W.
,
Xu
,
J.
,
Luo
,
X.
and
Zhang
,
Y.
(
2020
).
Diagnostic accuracy of quantitative fetal fibronectin to predict spontaneous preterm birth: a meta-analysis
.
Int. J. Gynaecol. Obstet.
153
,
220
-
227
.
Chi
,
F.
,
Jong
,
T. D.
,
Wang
,
L.
,
Ouyang
,
Y.
,
Wu
,
C.
,
Li
,
W.
and
Huang
,
S. H.
(
2010
).
Vimentin-mediated signalling is required for IbeA+ E. coli K1 invasion of human brain microvascular endothelial cells
.
Biochem. J.
427
,
79
-
90
.
Chi
,
F.
,
Bo
,
T.
,
Wu
,
C. H.
,
Jong
,
A.
and
Huang
,
S. H.
(
2012
).
Vimentin and PSF act in concert to regulate IbeA+ E. coli K1 induced activation and nuclear translocation of NF-κB in human brain endothelial cells
.
PLoS ONE
7
,
e35862
.
Chisholm
,
S. T.
,
Coaker
,
G.
,
Day
,
B.
and
Staskawicz
,
B. J.
(
2006
).
Host-microbe interactions: shaping the evolution of the plant immune response
.
Cell
124
,
803
-
814
.
Choi
,
Y. J.
,
Kim
,
N.
,
Chang
,
H.
,
Lee
,
H. S.
,
Park
,
S. M.
,
Park
,
J. H.
,
Shin
,
C. M.
,
Kim
,
J. M.
,
Kim
,
J. S.
,
Lee
,
D. H.
et al. 
(
2015
).
Helicobacter pylori-induced epithelial-mesenchymal transition, a potential role of gastric cancer initiation and an emergence of stem cells
.
Carcinogenesis
36
,
553
-
563
.
Coburn
,
J.
,
Dillon
,
S. T.
,
Iglewski
,
B. H.
and
Gill
,
D. M.
(
1989
).
Exoenzyme S of Pseudomonas aeruginosa ADP-ribosylates the intermediate filament protein vimentin
.
Infect. Immun.
57
,
996
-
998
.
Corsiero
,
E.
,
Bombardieri
,
M.
,
Carlotti
,
E.
,
Pratesi
,
F.
,
Robinson
,
W.
,
Migliorini
,
P.
and
Pitzalis
,
C.
(
2016
).
Single cell cloning and recombinant monoclonal antibodies generation from RA synovial B cells reveal frequent targeting of citrullinated histones of NETs
.
Ann. Rheum. Dis.
75
,
1866
-
1875
.
Coye
,
L. H.
and
Collins
,
C. M.
(
2004
).
Identification of SpyA, a novel ADP-ribosyltransferase of Streptococcus pyogenes
.
Mol. Microbiol.
54
,
89
-
98
.
Creasey
,
E. A.
and
Isberg
,
R. R.
(
2014
).
Maintenance of vacuole integrity by bacterial pathogens
.
Curr. Opin. Microbiol.
17
,
46
-
52
.
Daley
,
C. L.
(
2017
).
Mycobacterium avium Complex Disease
.
Microbiol. Spectr
.
5
,
microbiolspec.TNMI7-0045-2017
.
Deng
,
L.
,
Spencer
,
B. L.
,
Holmes
,
J. A.
,
Mu
,
R.
,
Rego
,
S.
,
Weston
,
T. A.
,
Hu
,
Y.
,
Sanches
,
G. F.
,
Yoon
,
S.
,
Park
,
N.
et al. 
(
2019
).
The group B streptococcal surface antigen I/II protein, BspC, interacts with host vimentin to promote adherence to brain endothelium and inflammation during the pathogenesis of meningitis
.
PLoS Pathog.
15
,
e1007848
.
Dos Santos
,
G.
,
Rogel
,
M. R.
,
Baker
,
M. A.
,
Troken
,
J. R.
,
Urich
,
D.
,
Morales-Nebreda
,
L.
,
Sennello
,
J. A.
,
Kutuzov
,
M. A.
,
Sitikov
,
A.
,
Davis
,
J. M.
et al. 
(
2015
).
Vimentin regulates activation of the NLRP3 inflammasome
.
Nat. Commun.
6
,
6574
.
Duncan-Lowey
,
J. K.
,
Wiscovitch
,
A. L.
,
Wood
,
T. E.
,
Goldberg
,
M. B.
and
Russo
,
B. C.
(
2020
).
Shigella flexneri disruption of cellular tension promotes intercellular spread
.
Cell Reports
33
,
108409
.
Dupont
,
A.
,
Tokarski
,
C.
,
Dekeyzer
,
O.
,
Guihot
,
A. L.
,
Amouyel
,
P.
,
Rolando
,
C.
and
Pinet
,
F.
(
2004
).
Two-dimensional maps and databases of the human macrophage proteome and secretome
.
Proteomics
4
,
1761
-
1778
.
Flitney
,
E. W.
,
Kuczmarski
,
E. R.
,
Adam
,
S. A.
and
Goldman
,
R. D.
(
2009
).
Insights into the mechanical properties of epithelial cells: the effects of shear stress on the assembly and remodeling of keratin intermediate filaments
.
FASEB J.
23
,
2110
-
2119
.
Fu
,
Y.
and
Galán
,
J. E.
(
1998
).
The Salmonella typhimurium tyrosine phosphatase SptP is translocated into host cells and disrupts the actin cytoskeleton
.
Mol. Microbiol.
27
,
359
-
368
.
Fu
,
A.
,
Yao
,
B.
,
Dong
,
T.
,
Chen
,
Y.
,
Yao
,
J.
,
Liu
,
Y.
,
Li
,
H.
,
Bai
,
H.
,
Liu
,
X.
,
Zhang
,
Y.
et al. 
(
2022
).
Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer
.
Cell
185
,
1356
-
1372.e26
.
Fuchs
,
E.
and
Weber
,
K.
(
1994
).
Intermediate filaments: structure, dynamics, function, and disease
.
Annu. Rev. Biochem..
63
,
345
-
382
.
Gasse
,
P.
,
Riteau
,
N.
,
Charron
,
S.
,
Girre
,
S.
,
Fick
,
L.
,
Pétrilli
,
V.
,
Tschopp
,
J.
,
Lagente
,
V.
,
Quesniaux
,
V. F. J.
,
Ryffel
,
B.
et al. 
(
2009
).
Uric acid is a danger signal activating NALP3 inflammasome in lung injury inflammation and fibrosis
.
Am. J. Respir. Crit. Care. Med.
179
,
903
-
913
.
Geisler
,
F.
and
Leube
,
R. E.
(
2016
).
Epithelial intermediate filaments: guardians against microbial infection?
Cells
5
,
29
.
Ghosh
,
P.
,
Halvorsen
,
E. M.
,
Ammendolia
,
D. A.
,
Mor-Vaknin
,
N.
,
O'riordan
,
M. X. D.
,
Brumell
,
J. H.
,
Markovitz
,
D. M.
and
Higgins
,
D. E.
(
2018
).
Invasion of the brain by listeria monocytogenes is mediated by InlF and host cell vimentin
.
mBio
9
,
e00160-18
.
Gladue
,
D. P.
,
O'donnell
,
V.
,
Baker-Branstetter
,
R.
,
Holinka
,
L. G.
,
Pacheco
,
J. M.
,
Fernández Sainz
,
I.
,
Lu
,
Z.
,
Ambroggio
,
X.
,
Rodriguez
,
L.
and
Borca
,
M. V.
(
2013
).
Foot-and-mouth disease virus modulates cellular vimentin for virus survival
.
J. Virol.
87
,
6794
-
6803
.
Gloire
,
G.
,
Legrand-Poels
,
S.
and
Piette
,
J.
(
2006
).
NF-κB activation by reactive oxygen species: Fifteen years later
.
Biochem. Pharmacol.
72
,
1493
-
1505
.
Guignot
,
J.
and
Servin
,
A. L.
(
2008
).
Maintenance of the Salmonella-containing vacuole in the juxtanuclear area: A role for intermediate filaments
.
Microb. Pathog.
45
,
415
-
422
.
Herrmann
,
H.
and
Aebi
,
U.
(
2016
).
Intermediate Filaments: Structure and Assembly
.
Cold Spring Harb Perspect Biol
8
,
a018242
.
Helmke
,
B. P.
,
Goldman
,
R. D.
and
Davies
,
P. F.
(
2000
).
Rapid displacement of vimentin intermediate filaments in living endothelial cells exposed to flow
.
Circ. Res.
86
,
745
-
752
.
Henderson
,
P.
and
Stevens
,
C.
(
2012
).
The role of autophagy in crohn's disease
.
Cells
1
,
492
-
519
.
Henderson
,
P.
,
Wilson
,
D. C.
,
Satsangi
,
J.
and
Stevens
,
C.
(
2012
).
A role for vimentin in Crohn disease
.
Autophagy
8
,
1695
-
1696
.
Hirschfeld
,
J.
,
White
,
P. C.
,
Milward
,
M. R.
,
Cooper
,
P. R.
and
Chapple
,
I. L. C.
(
2017
).
Modulation of neutrophil extracellular trap and reactive oxygen species release by periodontal bacteria
.
Infect. Immun.
85
,
e00297-17
.
Hofman
,
P.
and
Vouret-Craviari
,
V.
(
2012
).
Microbes-induced EMT at the crossroad of inflammation and cancer
.
Gut Microbes
3
,
176
-
185
.
Hooper
,
L. V.
,
Littman
,
D. R.
and
Macpherson
,
A. J.
(
2012
).
Interactions between the microbiota and the immune system
.
Science (New York, N.Y.)
336
,
1268
-
1273
.
Hu
,
J.
,
Li
,
Y.
,
Hao
,
Y.
,
Zheng
,
T.
,
Gupta
,
S. K.
,
Parada
,
G. A.
,
Wu
,
H.
,
Lin
,
S.
,
Wang
,
S.
,
Zhao
,
X.
et al. 
(
2019
).
High stretchability, strength, and toughness of living cells enabled by hyperelastic vimentin intermediate filaments
.
Proc. Natl. Acad. Sci. U.S.A.
116
,
17175
-
17180
.
Huang
,
J. Y.
,
Goers Sweeney
,
E.
,
Guillemin
,
K.
and
Amieva
,
M. R.
(
2017
).
Multiple Acid Sensors Control Helicobacter pylori Colonization of the Stomach
.
PLoS Pathog.
13
,
e1006118
.
Huang
,
S. H.
,
Stins
,
M. F.
and
Kim
,
K. S.
(
2000
).
Bacterial penetration across the blood-brain barrier during the development of neonatal meningitis
.
Microbes Infect.
2
,
1237
-
1244
.
Icenogle
,
L. M.
,
Hengel
,
S. M.
,
Coye
,
L. H.
,
Streifel
,
A.
,
Collins
,
C. M.
,
Goodlett
,
D. R.
and
Moseley
,
S. L.
(
2012
).
Molecular and biological characterization of streptococcal SpyA-mediated ADP-ribosylation of intermediate filament protein vimentin
.
J. Biol. Chem.
287
,
21481
-
21491
.
Ivaska
,
J.
,
Pallari
,
H. M.
,
Nevo
,
J.
and
Eriksson
,
J. E.
(
2007
).
Novel functions of vimentin in cell adhesion, migration, and signaling
.
Exp. Cell Res.
313
,
2050
-
2062
.
Khandpur
,
R.
,
Carmona-Rivera
,
C.
,
Vivekanandan-Giri
,
A.
,
Gizinski
,
A.
,
Yalavarthi
,
S.
,
Knight
,
J. S.
,
Friday
,
S.
,
Li
,
S.
,
Patel
,
R. M.
,
Subramanian
,
V.
et al. 
(
2013
).
NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis
.
Sci Transl Med
5
,
178ra140
.
Kidd
,
M. E.
,
Shumaker
,
D. K.
and
Ridge
,
K. M.
(
2014
).
The role of Vimentin intermediate filaments in the progression of lung cancer
.
Am. J. Respir. Cell Mol. Biol.
50
,
1
-
6
.
Kim
,
K. S.
(
2003
).
Pathogenesis of bacterial meningitis: From bacteraemia to neuronal injury
.
Nat. Rev. Neurosci.
4
,
376
-
385
.
Kim
,
M. J.
,
Romero
,
R.
,
Gervasi
,
M. T.
,
Kim
,
J.-S.
,
Yoo
,
W.
,
Lee
,
D.-C.
,
Mittal
,
P.
,
Erez
,
O.
,
Kusanovic
,
J. P.
,
Hassan
,
S. S.
et al. 
(
2009
).
Widespread microbial invasion of the chorioamniotic membranes is a consequence and not a cause of intra-amniotic infection
.
Lab. Invest.
89
,
924
-
936
.
Klareskog
,
L.
,
Amara
,
K.
and
Malmström
,
V.
(
2014
).
Adaptive immunity in rheumatoid arthritis: anticitrulline and other antibodies in the pathogenesis of rheumatoid arthritis
.
Curr. Opin. Rheumatol.
26
,
72
-
79
.
Kraehenbuhl
,
J. P.
and
Neutra
,
M. R.
(
2000
).
Epithelial M cells: Differentiation and function
.
Annu. Rev. Cell Dev. Biol.
16
,
301
-
332
.
Kraxner
,
J.
,
Lorenz
,
C.
,
Menzel
,
J.
,
Parfentev
,
I.
,
Silbern
,
I.
,
Denz
,
M.
,
Urlaub
,
H.
,
Schwappach
,
B.
and
Köster
,
S.
(
2021
).
Post-translational modifications soften vimentin intermediate filaments
.
Nanoscale
13
,
380
-
387
.
Kroemer
,
G.
,
Mariño
,
G.
and
Levine
,
B.
(
2010
).
Autophagy and the integrated stress response
.
Mol. Cell
40
,
280
-
293
.
Kroken
,
A. R.
,
Kumar
,
N. G.
,
Yahr
,
T. L.
,
Smith
,
B. E.
,
Nieto
,
V.
,
Horneman
,
H.
,
Evans
,
D. J.
and
Fleiszig
,
S. M. J.
(
2022
).
Exotoxin S secreted by internalized Pseudomonas aeruginosa delays lytic host cell death
.
PLoS Pathog.
18
,
e1010306
.
Krzysiek-Maczka
,
G.
,
Targosz
,
A.
,
Szczyrk
,
U.
,
Strzałka
,
M.
,
Sliwowski
,
Z.
,
Brzozowski
,
T.
,
Czyz
,
J.
and
Ptak-Belowska
,
A.
(
2018
).
Role of Helicobacter pylori infection in cancer-associated fibroblast-induced epithelial-mesenchymal transition in vitro
.
Helicobacter
23
,
e12538
.
Ku
,
N. O.
,
Azhar
,
S.
and
Bishr Omary
,
M.
(
2002
).
Keratin 8 phosphorylation by p38 kinase regulates cellular keratin filament reorganization. Modulation by a keratin 1–like disease-causing mutation
.
J. Biol. Chem.
277
,
10775
-
10782
.
Kumar
,
Y.
and
Valdivia
,
R. H.
(
2008
).
Actin and intermediate filaments stabilize the chlamydia trachomatis vacuole by forming dynamic structural scaffolds
.
Cell Host Microbe
4
,
159
-
169
.
Lam
,
F. W.
,
Da
,
Q.
,
Guillory
,
B.
and
Cruz
,
M. A.
(
2018
).
Recombinant human vimentin binds to p-selectin and blocks neutrophil capture and rolling on platelets and endothelium
.
J. Immunol.
200
,
1718
-
1726
.
Lamouille
,
S.
,
Xu
,
J.
and
Derynck
,
R.
(
2014
).
Molecular mechanisms of epithelial-mesenchymal transition
.
Nat. Rev. Mol. Cell Biol.
15
,
178
-
196
.
Lee
,
J.
,
Roberts
,
J. A. S.
,
Atanasova
,
K. R.
,
Chowdhury
,
N.
,
Han
,
K.
and
Yilmaz
,
Ö.
(
2017
).
Human primary epithelial cells acquire an epithelial-mesenchymal-transition phenotype during long-term infection by the oral opportunistic pathogen, Porphyromonas gingivalis
.
Front. Cell Infect. Microbiol.
7
,
493
.
Li
,
F. J.
,
Surolia
,
R.
,
Li
,
H.
,
Wang
,
Z.
,
Kulkarni
,
T.
,
Liu
,
G.
,
De Andrade
,
J. A.
,
Kass
,
D. J.
,
Thannickal
,
V. J.
,
Duncan
,
S. R.
et al. 
(
2017
).
Autoimmunity to vimentin is associated with outcomes of patients with idiopathic pulmonary fibrosis
.
J. Immunol.
199
,
1596
-
1605
.
Li
,
Z.
,
Paulin
,
D.
,
Lacolley
,
P.
,
Coletti
,
D.
and
Agbulut
,
O.
(
2020
).
Vimentin as a target for the treatment of COVID-19
.
BMJ Open Respir Res
7
,
e000623
.
Lin
,
A. E.
,
Beasley
,
F. C.
,
Keller
,
N.
,
Hollands
,
A.
,
Urbano
,
R.
,
Troemel
,
E. R.
,
Hoffman
,
H. M.
and
Nizet
,
V.
(
2015
).
A group a Streptococcus ADP-ribosyltransferase toxin stimulates a protective interleukin 1β-dependent macrophage immune response
.
mBio
6
,
e00133
.
Liou
,
J.-T.
,
Lee
,
C.-M.
,
Lin
,
Y.-C.
,
Chen
,
C.-Y.
,
Liao
,
C.-C.
,
Lee
,
H.-C.
and
Day
,
Y.-J.
(
2013
).
P-selectin is required for neutrophils and macrophage infiltration into injured site and contributes to generation of behavioral hypersensitivity following peripheral nerve injury in mice
.
Pain
154
,
2150
-
2159
.
Liu
,
S.
,
Huang
,
J.
,
Zhang
,
Y.
,
Liu
,
Y.
,
Zuo
,
S.
and
Li
,
R.
(
2019
).
MAP2K4 interacts with Vimentin to activate the PI3K/AKT pathway and promotes breast cancer pathogenesis
.
Aging (Albany NY)
11
,
10697
-
10710
.
Loske
,
P.
,
Boato
,
F.
,
Hendrix
,
S.
,
Piepgras
,
J.
,
Just
,
I.
,
Ahnert-Hilger
,
G.
and
Höltje
,
M.
(
2012
).
Minimal essential length of Clostridium botulinum C3 peptides to enhance neuronal regenerative growth and connectivity in a non-enzymatic mode
.
J. Neurochem.
120
,
1084
-
1096
.
Lowery
,
J.
,
Kuczmarski
,
E. R.
,
Herrmann
,
H.
and
Goldma
,
R. D.
(
2015
).
Intermediate filaments play a pivotal role in regulating cell architecture and function
.
J. Biol. Chem.
290
,
17145
-
17153
.
Luime
,
J. J.
,
Colin
,
E. M.
,
Hazes
,
J. M. W.
and
Lubberts
,
E.
(
2010
).
Does anti-mutated citrullinated vimentin have additional value as a serological marker in the diagnostic and prognostic investigation of patients with rheumatoid arthritis? A systematic review
.
Ann. Rheum. Dis.
69
,
337
-
344
.
Mahesh
,
P. P.
,
Retnakumar
,
R. J.
and
Mundayoor
,
S.
(
2016
).
Downregulation of vimentin in macrophages infected with live Mycobacterium tuberculosis is mediated by Reactive Oxygen Species
.
Sci. Rep.
6
,
21526
.
Mak
,
T. N.
,
Fischer
,
N.
,
Laube
,
B.
,
Brinkmann
,
V.
,
Metruccio
,
M. M. E.
,
Sfanos
,
K. S.
,
Mollenkopf
,
H. J.
,
Meyer
,
T. F.
and
Brüggemann
,
H.
(
2012
).
Propionibacterium acnes host cell tropism contributes to vimentin-mediated invasion and induction of inflammation
.
Cell. Microbiol.
14
,
1720
-
1733
.
Mak
,
T. N.
, and
Brüggemann
,
H.
(
2016
).
Vimentin in Bacterial Infections
.
Cells
5
,
18
.
Marques
,
M. S.
,
Melo
,
J.
,
Cavadas
,
B.
,
Mendes
,
N.
,
Pereira
,
L.
,
Carneiro
,
F.
,
Figueiredo
,
C.
and
Leite
,
M.
(
2018
).
Afadin downregulation by helicobacter pyloriInduces epithelial to mesenchymal transition in gastric cells
.
Front. Microbiol.
9
,
2712
.
Mcdonald-Hyman
,
C.
,
Muller
,
J. T.
,
Loschi
,
M.
,
Thangavelu
,
G.
,
Saha
,
A.
,
Kumari
,
S.
,
Reichenbach
,
D. K.
,
Smith
,
M. J.
,
Zhang
,
G.
,
Koehn
,
B. H.
et al. 
(
2018
).
The vimentin intermediate filament network restrains regulatory T cell suppression of graft-versus-host disease
.
J. Clin. Investig.
128
,
4604
-
4621
.
Mishra
,
B. B.
,
Moura-Alves
,
P.
,
Sonawane
,
A.
,
Hacohen
,
N.
,
Griffiths
,
G.
,
Moita
,
L. F.
and
Anes
,
E.
(
2010
).
Mycobacterium tuberculosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome
.
Cell Microbiol
.
12
,
1046
-
1063
.
Mitra
,
A.
,
Satelli
,
A.
,
Xia
,
X.
,
Cutrera
,
J.
,
Mishra
,
L.
and
Li
,
S.
(
2015
).
Cell-surface Vimentin: A mislocalized protein for isolating csVimentin+CD133- novel stem-like hepatocellular carcinoma cells expressing EMT markers
.
Int. J. Cancer
137
,
491
-
496
.
Mizushima
,
N.
and
Levine
,
B.
(
2020
).
Autophagy in human diseases
.
N. Engl. J. Med.
383
,
1564
-
1576
.
Moisan
,
E.
and
Girard
,
D.
(
2006
).
Cell surface expression of intermediate filament proteins vimentin and lamin B 1 in human neutrophil spontaneous apoptosis
.
J. Leukoc. Biol.
79
,
489
-
498
.
Mor-Vaknin
,
N.
,
Punturieri
,
A.
,
Sitwala
,
K.
and
Markovitz
,
D. M.
(
2003
).
Vimentin is secreted by activated macrophages
.
Nat. Cell Biol.
5
,
59
-
63
.
Mor-Vaknin
,
N.
,
Legendre
,
M.
,
Yu
,
Y.
,
Serezani
,
C. H. C.
,
Garg
,
S. K.
,
Jatzek
,
A.
,
Swanson
,
M. D.
,
Gonzalez-Hernandez
,
M. J.
,
Teitz-Tennenbaum
,
S.
,
Punturieri
,
A.
et al. 
(
2013
).
Murine colitis is mediated by Vimentin
.
Sci. Rep.
3
,
1045
.
Mortensen
,
J. H.
,
Godskesen
,
L. E.
,
Jensen
,
M. D.
,
Van Haaften
,
W. T.
,
Klinge
,
L. G.
,
Olinga
,
P.
,
Dijkstra
,
G.
,
Kjeldsen
,
J.
,
Karsdal
,
M. A.
,
Bay-Jensen
,
A. C.
et al. 
(
2015
).
Fragments of Citrullinated and MMP-degraded Vimentin and MMP-degraded Type III Collagen Are Novel Serological Biomarkers to Differentiate Crohn's Disease from Ulcerative Colitis
.
Journal of Crohn's & colitis
9
,
863
-
872
.
Murli
,
S.
,
Watson
,
R. O.
and
Gala
,
J. E.
(
2001
).
Role of tyrosine kinases and the tyrosine phosphatase SptP in the interaction of Salmonella with host cells
.
Cell. Microbiol.
3
,
795
-
810
.
Mutua
,
V.
and
Gershwin
,
L. J.
(
2021
).
A review of neutrophil extracellular traps (NETs) in disease: potential anti-NETs therapeutics
.
Clin. Rev. Allergy Immunol.
61
,
194
-
211
.
Nédellec
,
P.
,
Vicart
,
P.
,
Laurent-Winter
,
C.
,
Martinat
,
C.
,
Prévost
,
M.-C.
and
Brahic
,
M.
(
1998
).
Interaction of Theiler's virus with intermediate filaments of infected cells
.
J. Virol.
72
,
9553
-
9560
.
Nicu
,
E. A.
and
Loos
,
B. G.
(
2016
).
Polymorphonuclear neutrophils in periodontitis and their possible modulation as a therapeutic approach
.
Periodontol 2000
71
,
140
-
163
.
Packwood
,
K.
,
Drewe
,
E.
,
Staples
,
E.
,
Webster
,
D.
,
Witte
,
T.
,
Litzman
,
J.
,
Egner
,
W.
,
Sargur
,
R.
,
Sewell
,
W.
,
Lopez-Granados
,
E.
et al. 
(
2010
).
NOD2 polymorphisms in clinical phenotypes of common variable immunodeficiency disorders
.
Clin. Exp. Immunol.
161
,
536
-
541
.
Pagani
,
I.
,
Ghezzi
,
S.
,
Ulisse
,
A.
,
Rubio
,
A.
,
Turrini
,
F.
,
Garavaglia
,
E.
,
Candiani
,
M.
,
Castilletti
,
C.
,
Ippolito
,
G.
,
Poli
,
G.
et al. 
(
2017
).
Human endometrial stromal cells are highly permissive to productive infection by zika virus
.
Sci. Rep.
7
,
44286
.
Pan
,
P.
,
Su
,
L.
,
Wang
,
X.
,
Chai
,
W.
,
Liu
,
D.
,
Song
,
L.
and
Xie
,
L.
(
2021
).
Vimentin regulation of autophagy activation in lung fibroblasts in response to lipopolysaccharide exposure in vitro
.
Ann. Transl. Med.
9
,
304
-
304
.
Park
,
S. H.
,
Yoon
,
S. J.
,
Choi
,
S.
,
Kim
,
J. S.
,
Lee
,
M. S.
,
Lee
,
S. J.
,
Lee
,
S. H.
,
Min
,
J. K.
,
Son
,
M. Y.
,
Ryu
,
C. M.
et al. 
(
2020
).
Bacterial type III effector protein HopQ inhibits melanoma motility through autophagic degradation of vimentin
.
Cell Death Dis.
11
,
231
.
Patteson
,
A. E.
,
Vahabikashi
,
A.
,
Pogoda
,
K.
,
Adam
,
S. A.
,
Mandal
,
K.
,
Kittisopikul
,
M.
,
Sivagurunathan
,
S.
,
Goldman
,
A.
,
Goldman
,
R. D.
and
Janmey
,
P. A.
(
2019
).
Vimentin protects cells against nuclear rupture and DNA damage during migration
.
J. Cell Biol.
218
,
4079
-
4092
.
Patteson
,
A. E.
,
Carroll
,
R. J.
,
Iwamoto
,
D. V.
and
Janmey
,
P. A.
(
2020a
).
The vimentin cytoskeleton: when polymer physics meets cell biology
.
Phys. Biol.
18
,
011001
.
Patteson
,
A. E.
,
Vahabikashi
,
A.
,
Goldman
,
R. D.
and
Janmey
,
P. A.
(
2020b
).
Mechanical and Non-mechanical functions of filamentous and non-filamentous vimentin
.
BioEssays
42
,
e2000078
.
Perry
,
A.
and
Lambert
,
P.
(
2011
).
Propionibacterium acnes: Infection beyond the skin
.
Expert Rev. Anti Infect. Ther.
9
,
1149
-
1156
.
Pinaud
,
L.
,
Sansonetti
,
P. J.
and
Phalipon
,
A.
(
2018
).
Host cell targeting by enteropathogenic bacteria T3SS effectors
.
Trends Microbiol.
26
,
266
-
283
.
Puhar
,
A.
and
Sansonetti
,
P. J.
(
2014
).
Type III secretion system
.
Curr. Biol.
24
,
R784
-
R791
.
Putz
,
K.
,
Hayani
,
K.
and
Zar
,
F. A.
(
2013
).
Meningitis
.
Prim. Care
40
,
707
-
726
.
Raabe
,
V. N.
and
Shane
,
A. L.
(
2019
).
Group B Streptococcus (Streptococcus agalactiae)
.
Microbiol. Spectr.
7
,
1
.
Ramos-Morales
,
F.
(
2012
).
Impact of salmonella enterica Type III secretion system effectors on the eukaryotic host cell
.
ISRN Cell Biol.
2012
,
1
-
36
.
Rivera
,
A.
(
2019
).
Interferon Lambda's new role as regulator of neutrophil function
.
J. Interferon Cytokine Res.
39
,
609
-
617
.
Russo
,
B. C.
,
Stamm
,
L. M.
,
Raaben
,
M.
,
Kim
,
C. M.
,
Kahoud
,
E.
,
Robinson
,
L. R.
,
Bose
,
S.
,
Queiroz
,
A. L.
,
Herrera
,
B. B.
,
Baxt
,
L. A.
et al. 
(
2016
).
Intermediate filaments enable pathogen docking to trigger type 3 effector translocation
.
Nat. Microbiol.
1
,
16025
-
16025
.
Satelli
,
A.
and
Li
,
S.
(
2011
).
Vimentin in cancer and its potential as a molecular target for cancer therapy
.
Cell. Mol. Life Sci.
68
,
3033
-
3046
.
Segawa
,
M.
,
Niino
,
K.
,
Mineki
,
R.
,
Kaga
,
N.
,
Murayama
,
K.
,
Sugimoto
,
K.
,
Watanabe
,
Y.
,
Furukawa
,
K.
and
Horigome
,
T.
(
2005
).
Proteome analysis of a rat liver nuclear insoluble protein fraction and localization of a novel protein, ISP36, to compartments in the interchromatin space
.
FEBS J.
272
,
4327
-
4338
.
Sharpe
,
A. H.
,
Chen
,
L. B.
,
Murphy
,
J. R.
and
Fields
,
B. N.
(
1980
).
Specific disruption of vimentin filament organization in monkey kidney CV-1 cells by diphtheria toxin, exotoxin A, and cycloheximide
.
Proc. Natl. Acad. Sci. U.S.A.
77
,
7267
-
7271
.
Snider
,
N. T.
and
Omary
,
M. B.
(
2014
).
Post-translational modifications of intermediate filament proteins: Mechanisms and functions
.
Nat. Rev. Mol. Cell Biol.
15
,
163
-
177
.
Snider
,
N. T.
,
Ku
,
N.-O.
and
Omary
,
M. B.
(
2018
).
The sweet side of vimentin
.
ELife
7
,
e35336
.
Steinert
,
P. M.
and
Liem
,
R. K.
(
1990
).
Intermediate filament dynamics
.
Cell
60
,
521
-
523
.
Stevens
,
C.
,
Henderson
,
P.
,
Nimmo
,
E. R.
,
Soares
,
D. C.
,
Dogan
,
B.
,
Simpson
,
K. W.
,
Barrett
,
J. C.
,
Wilson
,
D. C.
and
Satsangi
,
J.
(
2013
).
The intermediate filament protein, vimentin, is a regulator of NOD2 activity
.
Gut
62
,
695
-
707
.
Stuen
,
S.
,
Granquist
,
E. G.
and
Silaghi
,
C.
(
2013
).
Anaplasma phagocytophilum--a widespread multi-host pathogen with highly adaptive strategies
.
Front. Cell Infect. Microbiol.
3
,
31
.
Su
,
L.
,
Pan
,
P.
,
Yan
,
P.
,
Long
,
Y.
,
Zhou
,
X.
,
Wang
,
X.
,
Zhou
,
R.
,
Wen
,
B.
,
Xie
,
L.
and
Liu
,
D.
(
2019a
).
Role of vimentin in modulating immune cell apoptosis and inflammatory responses in sepsis
.
Sci. Rep.
9
,
5747
.
Su
,
L. X.
,
Pan
,
P.
,
Wang
,
X. T.
,
Long
,
Y.
,
Liu
,
D. W.
and
Zhou
,
X.
(
2019b
).
Vimentin modulates apoptosis and inflammatory cytokine release by a human monocytic cell line (THP-1) in response to lipopolysaccharides in vitro
.
Chinese Med. J.
132
,
1336
-
1343
.
Sukumaran
,
B.
,
Mastronunzio
,
J. E.
,
Narasimhan
,
S.
,
Fankhauser
,
S.
,
Uchil
,
P. D.
,
Levy
,
R.
,
Graham
,
M.
,
Colpitts
,
T. M.
,
Lesser
,
C. F.
and
Fikrig
,
E.
(
2011
).
Anaplasma phagocytophilum AptA modulates Erk1/2 signalling
.
Cell. Microbiol.
13
,
47
-
61
.
Sukumaran
,
A.
,
Woroszchuk
,
E.
,
Ross
,
T.
and
Geddes-Mcalister
,
J.
(
2021
).
Proteomics of host-bacterial interactions: new insights from dual perspectives
.
Can. J. Microbiol.
67
,
213
-
225
.
Tahoun
,
A.
,
Siszler
,
G.
,
Spears
,
K.
,
Mcateer
,
S.
,
Tree
,
J.
,
Paxton
,
E.
,
Gillespie
,
T. L.
,
Martinez-Argudo
,
I.
,
Jepson
,
M. A.
,
Shaw
,
D. J.
et al. 
(
2011
).
Comparative analysis of EspF variants in inhibition of Escherichia coli phagocytosis by macrophages and inhibition of E. coli translocation through human- and bovine-derived M cells
.
Infect. Immun.
79
,
4716
-
4729
.
Tahoun
,
A.
,
Mahajan
,
S.
,
Paxton
,
E.
,
Malterer
,
G.
,
Donaldson
,
D. S.
,
Wang
,
D.
,
Tan
,
A.
,
Gillespie
,
T. L.
,
O'shea
,
M.
,
Roe
,
A. J.
et al. 
(
2012
).
Salmonella transforms follicle-associated epithelial cells into M cells to promote intestinal invasion
.
Cell Host Microbe
12
,
645
-
656
.
Takeuchi
,
O.
and
Akira
,
S.
(
2010
).
Pattern recognition receptors and inflammation
.
Cell
140
,
805
-
820
.
Tarbet
,
H. J.
,
Dolat
,
L.
,
Smith
,
T. J.
,
Condon
,
B. M.
,
O'brien
,
E. T.
,
Valdivia
,
R. H.
and
Boyce
,
M.
(
2018
).
Site-specific glycosylation regulates the form and function of the intermediate filament cytoskeleton
.
eLife
7
,
e31807
.
Thanaphongdecha
,
P.
,
Karinshak
,
S. E.
,
Ittiprasert
,
W.
,
Mann
,
V. H.
,
Chamgramol
,
Y.
,
Pairojkul
,
C.
,
Fox
,
J. G.
,
Suttiprapa
,
S.
,
Sripa
,
B.
and
Brindley
,
P. J.
(
2020
).
Infection with helicobacter pylori induces epithelial to mesenchymal transition in human cholangiocytes
.
Pathogens
9
,
971
.
Thiagarajan
,
P. S.
,
Yakubenko
,
V. P.
,
Elsori
,
D. H.
,
Yadav
,
S. P.
,
Willard
,
B.
,
Tan
,
C. D.
,
René Rodriguez
,
E.
,
Febbraio
,
M.
and
Cathcart
,
M. K.
(
2013
).
Vimentin is an endogenous ligand for the pattern recognition receptor Dectin-1
.
Cardiovasc. Res.
99
,
494
-
504
.
Toivola
,
D. M.
,
Tao
,
G. Z.
,
Habtezion
,
A.
,
Liao
,
J.
and
Omary
,
M. B.
(
2005
).
Cellular integrity plus: Organelle-related and protein-targeting functions of intermediate filaments
.
Trends Cell Biol.
15
,
608
-
617
.
Truchan
,
H. K.
,
Cockburn
,
C. L.
,
May
,
L. J.
,
Viebrock
,
L.
and
Carlyon
,
J. A.
(
2016
).
Anaplasma phagocytophilum-occupied vacuole interactions with the host cell cytoskeleton
.
Veterinary Sciences
3
,
25
.
Tsolis
,
R. M.
,
Kingsley
,
R. A.
,
Townsend
,
S. M.
,
Ficht
,
T. A.
,
Adams
,
L. G.
and
Bäumler
,
A. J.
(
2000
).
Of mice, calves, and men: comparison of the mouse typhoid model with other Salmonella infections
.
Adv. Exp. Med. Biol.
473
,
261
-
274
.
Van Bodegraven
,
E. J.
and
Etienne-Manneville
,
S.
(
2021
).
Intermediate filaments from tissue integrity to single molecule mechanics
.
Cells
10
,
1905
.
Van Der Flier
,
M.
(
2021
).
Neonatal meningitis: small babies, big problem
.
Lancet Child Adolesc. Health
5
,
386
-
387
.
Van Engeland
,
N. C. A.
,
Suarez Rodriguez
,
F.
,
Rivero-Müller
,
A.
,
Ristori
,
T.
,
Duran
,
C. L.
,
Stassen
,
O. M. J. A.
,
Antfolk
,
D.
,
Driessen
,
R. C. H.
,
Ruohonen
,
S.
,
Ruohonen
,
S. T.
et al. 
(
2019
).
Vimentin regulates Notch signaling strength and arterial remodeling in response to hemodynamic stress
.
Sci. Rep.
9
,
12415
.
Van Rees
,
D. J.
,
Szilagyi
,
K.
,
Kuijpers
,
T. W.
,
Matlung
,
H. L.
and
Van Den Berg
,
T. K.
(
2016
).
Immunoreceptors on neutrophils
.
Semin. Immunol.
28
,
94
-
108
.
Van Steendam
,
K.
,
Tilleman
,
K.
and
Deforce
,
D.
(
2011
).
The relevance of citrullinated vimentin in the production of antibodies against citrullinated proteins and the pathogenesis of rheumatoid arthritis
.
Rheumatology (Oxf.)
50
,
830
-
837
.
Vossenaar
,
E. R.
,
Radstake
,
T. R. D.
,
Van Der Heijden
,
A.
,
Van Mansum
,
M. A. M.
,
Dieteren
,
C.
,
De Rooij
,
D. J.
,
Barrera
,
P.
,
Zendman
,
A. J. W.
and
Van Venrooij
,
W. J.
(
2004
).
Expression and activity of citrullinating peptidylarginine deiminase enzymes in monocytes and macrophages
.
Ann. Rheum. Dis.
63
,
373
-
381
.
Wroblewski
,
L. E.
,
Piazuelo
,
M. B.
,
Chaturvedi
,
R.
,
Schumacher
,
M.
,
Aihara
,
E.
,
Feng
,
R.
,
Noto
,
J. M.
,
Delgado
,
A.
,
Israel
,
D. A.
,
Zavros
,
Y.
et al. 
(
2015
).
Helicobacter pylori targets cancer-associated apical-junctional constituents in gastroids and gastric epithelial cells
.
Gut
64
,
720
-
730
.
Xiong
,
Q.
and
Rikihisa
,
Y.
(
2011
).
The prenylation inhibitor manumycin A reduces the viability of Anaplasma phagocytophilum
.
J. Med. Microbiol.
60
,
744
-
749
.
Xiong
,
Q.
,
Lin
,
M.
and
Rikihisa
,
Y.
(
2009
).
Cholesterol-dependent anaplasma phagocytophilum exploits the low-density lipoprotein uptake pathway
.
PLoS Pathog.
5
,
e1000329
.
Yago
,
T.
,
Leppänen
,
A.
,
Carlyon
,
J. A.
,
Akkoyunlu
,
M.
,
Karmakar
,
S.
,
Fikrig
,
E.
,
Cummings
,
R. D.
and
Mcever
,
R. P.
(
2003
).
Structurally distinct requirements for binding of P-selectin glycoprotein ligand-1 and sialyl Lewis x to Anaplasma phagocytophilum and P-selectin
.
J. Biol. Chem.
278
,
37987
-
37997
.
Yang
,
Y.
and
Zhang
,
C. Y.
(
2014
).
Visualizing and quantifying protein polySUMOylation at the single-molecule level
.
Anal. Chem.
86
,
967
-
972
.
Yang
,
Y.
,
Hu
,
M.
,
Yu
,
K.
,
Zeng
,
X.
and
Liu
,
X.
(
2015
).
Mass spectrometry-based proteomic approaches to study pathogenic bacteria-host interactions
.
Protein Cell
6
,
265
-
274
.
Yu
,
H.
,
Zeng
,
J.
,
Liang
,
X.
,
Wang
,
W.
,
Zhou
,
Y.
,
Sun
,
Y.
,
Liu
,
S.
,
Li
,
W.
,
Chen
,
C.
and
Jia
,
J.
(
2014
).
Helicobacter pylori promotes epithelial-mesenchymal transition in gastric cancer by Downregulating Programmed Cell Death Protein 4 (PDCD4)
.
PLoS ONE
9
,
e105306
.
Yu
,
M. B.
,
Guerra
,
J.
,
Firek
,
A.
and
Langridge
,
W. H. R.
(
2018
).
Extracellular vimentin modulates human dendritic cell activation
.
Mol. Immunol.
104
,
37
-
46
.
Zhang
,
X.
,
Shi
,
H. Y.
,
Chen
,
J. F.
,
Shi
,
D.
,
Dong
,
H.
and
Feng
,
L.
(
2015
).
Identification of the interaction between vimentin and nucleocapsid protein of transmissible gastroenteritis virus
.
Virus Res.
200
,
56
-
63
.
Zhang
,
Y.
,
Wen
,
Z.
,
Shi
,
X.
,
Liu
,
Y. J.
,
Eriksson
,
J. E.
and
Jiu
,
Y.
(
2021
).
The diverse roles and dynamic rearrangement of vimentin during viral infection
.
J. Cell Sci.
134
,
jcs250597
.
Zhang
,
Y.
,
Zhao
,
S.
,
Li
,
Y.
,
Feng
,
F.
,
Li
,
M.
,
Xue
,
Y.
,
Cui
,
J.
,
Xu
,
T.
,
Jin
,
X.
and
Jiu
,
Y.
(
2022
).
Host cytoskeletal vimentin serves as a structural organizer and an RNA-binding protein regulator to facilitate Zika viral replication
.
Proc. Natl. Acad. Sci. USA
119
,
e2113909119
.
Zhou
,
D.
and
Galán
,
J.
(
2001
).
Salmonella entry into host cells: the work in concert of type III secreted effector proteins
.
Microbes Infect.
3
,
1293
-
1298
.
Zhou
,
H.-F.
,
Ren
,
K.
,
Zha
,
J.-H.
,
Guo
,
Q.
and
Zhao
,
G.-J.
(
2020
).
Vimentin promotes endothelial inflammation by activating NLRP3
.
Int. J. Cardiol.
301
,
155
.
Zou
,
Y.
,
He
,
L.
and
Huang
,
S. H.
(
2006
).
Identification of a surface protein on human brain microvascular endothelial cells as vimentin interacting with Escherichia coli invasion protein IbeA
.
Biochem. Biophys. Res. Commun.
351
,
625
-
630
.

Competing interests

The authors declare no competing or financial interests.