Abstract
The passive and regulated movement of ions, solutes, and water via spaces between cells of the epithelial monolayer plays a critical role in the normal intestinal functioning. This paracellular pathway displays a high level of structural and functional specialization, with the membrane-spanning complexes of the tight junctions, adherens junctions, and desmosomes ensuring its integrity. Tight junction proteins, like occludin, tricellulin, and the claudin family isoforms, play prominent roles as barriers to unrestricted paracellular transport. The past decade has witnessed major advances in our understanding of the architecture and function of epithelial tight junctions. While it has been long appreciated that microbes, notably bacterial and viral pathogens, target and disrupt junctional complexes and alter paracellular permeability, the precise mechanisms remain to be defined. Notably, renewed efforts will be required to interpret the available data on pathogen-mediated barrier disruption in the context of the most recent findings on tight junction structure and function. While much of the focus has been on pathogen-induced dysregulation of junctional complexes, commensal microbiota and their products may influence paracellular permeability and contribute to the normal physiology of the gut. Finally, microbes and their products have become important tools in exploring host systems, including the junctional properties of epithelial cells.
Introduction
The gut epithelium, with the exception of the stratified layers in the esophagus, is a single contiguous layer of cells that represents a unique barrier between the diverse luminal contents and the underlying tissue (67). The epithelium is vested with the exacting task of processing food, absorbing water and nutrition, and compacting and disposing undigested materials while, simultaneously, keeping pathogens and toxic molecules at bay. Paralleling the discrete tasks along its length, the epithelial cell composition and character, as well as the substances secreted or absorbed, differ in the various segments of the gut. Concomitantly, these niches, with distinct pH, oxygen levels, and nutrients, are home to diverse arrays of microbes. The microbial communities that thrive in these niches also intimately influence the biology of the gut and beyond. Importantly, they often bolster host defenses by preventing the colonization of pathogenic organisms, a feature termed as colonization resistance (52).
The epithelial cells of the gut achieve their unique tasks by maintaining apical-basal polarity (108). This is essential for the directional transport of nutrients and water, as well as the secretion of compounds such as antimicrobial peptides and enzymes into the lumen. The various junctional complexes between adjacent epithelial cells serve many functions. Most fundamentally, they tether the cells together as a monolayer, and provide mechanical strength to withstand the passage of food and water. Beyond this, they contribute to the “fence” function, that is, the maintenance of cell polarity by preventing lateral diffusion of membrane molecules between the apical and basolateral sides. Finally, they regulate the passive transport of water, ions, and molecules along the paracellular pathway, a property known as barrier function.
The apical-most tight junction (TJ) complexes are the major players in regulating the fence and barrier functions (133). Mechanical strength for the monolayer is provided by the more basal desmosomes, the “spot-welds” that hold epithelial cells together. Other junctional complexes include the adherens junctions (AJs) and gap junctions, which play various roles including the coordination of membrane events with intracellular signaling changes. The junctional complexes are variously tethered to the intracellular cytoskeletal elements, namely the actin microfilaments, microtubules, and intermediate filaments.
The organization and function of the epithelium needs to be dynamically maintained despite exposure to an array of substances and organisms in the lumen (133). In the normal intestine, there are many opportunities for breaches in the monolayer, such as luminal sampling of antigens by underlying immune cells, wounds and wound healing, programmed cell death, the extrusion of dead cells, and cell proliferation and replenishment. In relation to the last point, it is worth noting that the entire lining of the intestine renews itself once every 4 to 8 days in the adult gut. Discrete processes operate to ensure that the integrity of the paracellular pathway, and the polarity of the epithelial cells, is maintained despite these perturbations.
Polymorphisms and mutations in TJ proteins have been linked to diseases of various tissues and organs (133). For instance, claudin mutations are associated with ichthyosis (claudin-1), velo-cardial facial syndrome (claudin-5), nonsyndromic deafness (claudin-14), familial hypomagnesaemia (claudin-16 and −19), and vision loss (claudin-19), and tricellulin mutations are associated with nonsyndromic deafness. Intestinal TJ protein expression and distribution is altered in many pathologic states including inflammatory bowel disease (37). Many pathogens perturb junctional complexes and dysregulate paracellular permeability, which contributes to disease. Bacteria and viruses may bind to cell junctions, or recruit junctional proteins to regions of attachment. Often, pathogens dislocate TJ proteins from the apical junctional regions, causing their movement down along the lateral membrane, and to intracellular locations. Pathogen-mediated signaling alterations can cause contraction of the perijunctional ring of actin and increase paracellular permeability.
This overview article reviews the mechanisms by which select pathogens target intestinal epithelial TJ complexes and alter paracellular permeability. To provide context, a brief summary of the normal structure and function of TJs, and their role in regulating paracellular permeability, is included. Turner and Turner provide a comprehensive picture of the gastrointestinal tract (133), while the overview article by Madara (67) provides a review on epithelial morphology in the small intestine. For excellent overviews on TJ structure and function, the reader is referred to other recent articles (33, 155).
Overview of Cell Junctional Complexes
The apical junctional complex (AJC) is a highly organized structure located at the apex of the lateral membrane of polarized epithelial cells (97). The AJC is involved in cell-cell adhesion and paracellular permeability and, by separating the apical and basolateral domains, maintains epithelial cell polarity. It is comprised of the apical-most TJs, followed by the AJs and desmosomes. While these structures have unique compositions with relatively few common proteins, they share some overall features. Each junctional complex is composed of integral membrane proteins that form intercellular bridges, adaptor proteins that anchor the membrane components to cytoskeletal structures, and specific signaling molecules (155).
Thus, the TJ complex includes transmembrane proteins of the claudin family that mediate interactions between adjacent cells, and subjunctional adaptor proteins like cingulin and members of the zona occludens family. The protein-protein interaction domains of adaptor proteins help tether the membrane proteins to microtubules and actin filaments. Below the TJs are the AJs, containing the membrane-spanning classical E-cadherins and nectins, the cytosolic interaction proteins α- and β-catenin and p120 catenin (for E-cadherin) and afadin (for nectins). AJs are required for the assembly of the paracellular barrier and the regulation of mucosal permeability (60). Below the TJ and AJ are the desmosomes, the “spot-welds” that provide mechanical integrity to the epithelial monolayer. Desmosomes are composed of the transmembrane desmosomal cadherins desmoglein and desmocollin, and associated cytosolic plaque proteins plakophilin, plakoglobin, and desmoplakin. Desmoplakin anchors the desmosomal complex to the keratin intermediate filament network. Emerging studies suggest a role for the desmosomal junctions in potentiating epithelial barrier function (118, 137).
Structural features of tight junctions
In electron micrographs, TJs appear as focal “kissing” contacts between neighboring columnar cells, supported by electron-dense cytoplasmic plaques (67). Freeze fracture micrography, a technique that provides planar views of the hydrophobic interior of membranes, shows the TJ membrane proteins as an anastomosing meshwork of strands (155). Broadly, the number of strands correlates positively with the leakiness of the epithelium; fewer strands are present in the “leaky” epithelia of the gut compared to tight epithelia such as those of the urinary bladder (19). However, the occurrence of epithelial systems with similar number of strands, but vastly different transepithelial resistances, suggests that TJ composition also dictates barrier properties (36, 41, 120).
The TJ strands or fibrils are composed of tetraspan transmembrane proteins of the claudin family (26 isoforms in humans) and members of the TJ-associated MARVEL [myelin and lymphocyte protein (MAL) and related proteins for vesicle trafficking and membrane link] proteins, occludin, tricellulin, and MARVELD3. Occludin is primarily localized to junctions between two neighboring cells (bicellular junctions), while tricellulin and anguillins are enriched at the points of contact of three neighboring cells (tricellular junctions). Additional components localizing to the TJs include the single-span transmembrane proteins junction-adhesion molecules (JAMs) and coxsackievirus- and adenovirus-receptor (CAR), which contain immunoglobulin-like domains.
Cytosolic TJ-associated proteins, such as the zona occludens proteins ZO-1, ZO-2, and ZO-3, and cingulin, contain various protein-protein interaction motifs that facilitate their adaptor function. ZO-1, the first identified TJ protein, as well as the best studied to date, harbors N-terminal domains that facilitate interaction with the membrane-anchored claudins, occludin, and JAM, and with the transcriptional regulator ZO-1–associated nucleic acid binding proteins (ZONAB) and heat-shock protein, APG2; the C-terminal half of ZO-1 interacts with F-actin. Cytoplasmic plaque proteins may have an ordered structure, and knockout/knockdown studies suggest some level of redundancy. Also closely associated with the TJs are partitioning complexes that play a role in generating and maintaining the apical-basal polarity of epithelial cells (99). Signaling molecules localizing to the TJs, such as the small GTPases RhoA, Rac, and CDC42 and their regulators, and atypical protein kinase C (aPKC), modulate epithelial barrier function. Finally, lipids are recognized to be fundamentally important for TJ structure and function, but their precise role in the maintenance of barrier function is poorly understood (155).
Modes of paracellular permeability
The main gatekeepers of paracellular transport are TJs, which have the added role of limiting the lateral movement of membrane-associated molecules and, thereby, maintaining the polarity of the epithelial monolayer (“gate” and “fence” functions, respectively). In the simple epithelia of the gut, TJs are dynamic structures that form a semipermeable diffusion barrier to ions, solutes, and water, while restricting the passage of bacteria and toxins.
Two modes of paracellular transport, the pore and leak pathways, can be discerned (106, 138, 142). The high-capacity pore pathway is charge- and size-selective, accommodating molecules ranging from 5 to 10 Å (54, 126). The leak pathway, which has limited capacity, is not selective for charge and facilitates transport of larger molecules, with one study estimating a sieving radius of ~62.5 Å (12). Distinct mechanisms govern the pore and leak pathways, and they can be independently, even opposingly, regulated. In addition to these pathways, epithelial damage can allow mass movement of small and large molecules, as well as microbes and toxins. Movement via this unrestricted pathway, not strictly a mode of paracellular transport, may play a prominent role in the context of microbial pathogenesis, particularly during later stages of infection.
Claudins are the key determinants of TJ permeability properties (37). Claudin isoforms may offer cation-selectivity (claudin-2, −10b, and −15), anion-selectivity (claudin-10a and −17), or enhance the sealing or barrier function of TJs (claudin-1, −4, and −8). The first two extracellular loop of claudins influence ion selectivity. One proposed mechanism for the action of barrier enhancing claudins is via the inhibition of the function of pore-forming claudins. Proteins of the MARVEL family can impact TJ strand architecture and regulate claudin channel function. For instance, occludin phosphorylation status influences claudin-2 localization at the TJs and, consequently, paracellular cation flux (98).
Regulation of TJ barrier function by the perijunctional actomyosin ring
Actin filaments within the microvilli of epithelial cells descend into the terminal web, a meshwork containing actin, actin-binding proteins, myosin, and intermediate filaments. At the cell peripheries, the terminal web condenses into a dense ring of actin at the region of the adherens and TJs (23). Filamentous actin associates with nonmuscle myosin II (NM II), which is composed of heavy chains, essential light chains, and regulatory light chains. Pharmacological inhibition of NM II by blebbistatin, or siRNA-mediated depletion of this molecule, resulted in a loss of barrier function despite apparent structural integrity of the junctional complexes (97). Mice with intestinal epithelial-specific knockout of the NM II heavy chain had altered expression/localization of junctional proteins, and increased intestinal permeability (86). Various mediators of barrier disruption, including calcium depletion and proinflammatory cytokines, activate NM II.
Phosphorylation of regulatory myosin light chain (MLC) results in unfolding of the actin-binding domain, and contraction of the peripheral actomyosin ring (97). This leads to TJ opening and, consequently, an increase in paracellular permeability. The phosphorylation status of MLC is dictated by several proteins including Rho GTPases, Rho-associated protein kinase (ROCK), MLC kinase (MLCK) and MLC phosphatase. Apart from directly phosphorylating MLC, ROCK is also an inhibitor of MLC phosphatase. Altered paracellular permeability resulting from actomyosin contraction has a normal physiological role in the intestine. For instance, activation of transcellular Na+/glucose cotransport by the apical transporter SGLT1 results in MLCK activation and MLC phosphorylation (135). The resulting TJ opening promotes paracellular absorption of water and small nutrients (radius <3.6 Å) along the osmotic gradient. This normal physiological pathway underlies the success of oral rehydration solutions containing glucose and sodium chloride in treating diarrhea. In a pathological context, several intestinal pathogens cause diarrhea partly via the phosphorylation and activation of myosin II.
The junction-associated actin cytoskeleton undergoes constant turnover, and molecules that regulate actin dynamics, such as actin-depolymerization factor (ADF)/cofilin and actin-interacting protein 1 (AIP1), regulate the assembly and permeability of the epithelial barrier (59, 141). More recently, the actin-interacting molecule cortactin was implicated in epithelial barrier function regulation (18). Cortactin deficiency enhanced RhoA/ROCK-mediated actomyosin contraction, and ROCK1 inhibition restored barrier function. Multiple lines of evidence indicate that the maintenance of epithelial barrier integrity requires the balanced regulation of Rho GTPase activity: both the excess and depletion of active Rho GTPases can perturb paracellular permeability (60).
Assays for measuring paracellular permeability
Several complementary methods are used to test paracellular permeability (106). The most commonly used method is the measurement of transepithelial electrical resistance (TER) to assess the flux of all ions across the epithelium. A current is applied across the epithelial monolayer, the generated potential is measured, and Ohm’s law is used to calculate the resistance to the flow of current. Since the small ions Na+ and Cl− are typically used in physiological solutions, TER measurements do not distinguish between the pore and leak pathways. Leak pathway permeability can be measured by quantitating the flux of charged or uncharged macromolecules of various sizes across the epithelium. Commonly used tracers include mannitol (molecular radius = 3.9 Å), lactulose (5.1 Å), inulin (10 Å), and varying sizes of polymers of polyethylene glycol (PEG 400 = 5.5 Å, PEG 4000 = 15.9 Å) or dextran (4 kDa = 4.9 Å, 40 kDa = 45 Å) (1, 39, 143). Charge and size selectivity of the pore pathway can be determined via dilution potential measurement and biionic substitution methods, respectively. While epithelial monolayers are known to display heterogeneity, with areas of high and low permeability (79), most measurement strategies focus on population averages. Recently, however, Weber et al. were able to use a novel patch-clamp technique to measure flux across individual claudin-2 channels (144). Most studies assessing the impact of microbes on paracellular permeability have involved TER measurements or tracer flux assays; pore selectivity measurements have seldom been performed.
Microbial Targeting of Junctional Components and Regulation of Paracellular Permeability
Many pathogenic bacteria and viruses target junctional complexes and perturb epithelial barrier function. Pathogens or their toxins may use junctional proteins as receptors. For instance, claudin family proteins serve as receptors for hepatitis C virus (HCV), as well as the Clostridium perfringens enterotoxin (21, 109). Even organisms like enterohemorrhagic Escherichia coli, which do not have an identified junctional receptor, line up along the junctions of epithelial cells (“log jam” phenotype) (76). Toxins like the Vibrio cholerae hemagglutinin/protease (metalloprotease) and the pore-forming aerolysin of Aeromonas hydrophila can directly cleave the TJ protein occludin (85).
Diverse microbes alter intracellular signaling events, leading to the posttranslational modification and displacement of junctional proteins (Fig. 1) (27). Pathogens such as enteropathogenic E. coli and Helicobacter pylori target epithelial cell polarity complexes, perturb apical-basal polarity, and alter trafficking of TJ proteins (Fig. 2) (128). In other instances, pathogen-induced changes in TJ protein expression can alter barrier function (77, 154). As an example, Salmonella causes the elevation of claudin-2, but not claudin3 (or claudin-7) in the colonic epithelial cells of infected mice (Fig. 3). Finally, microbe-mediated cytoskeletal changes, including MLC phosphorylation, can trigger actomyosin contraction and, thereby, alter paracellular permeability (Fig. 4).
Pathogen-induced inflammation may also be an indirect pathway to TJ alterations and barrier perturbation (60). For instance, many infectious agents trigger release of the proinflammatory molecule tumor necrosis factor α (TNFα). TNFα can stimulate leak pathway permeability by activating MLCK (Fig. 4) and inducing occludin endocytosis (134). Typically, microbe-induced permeability alterations are mediated via multiple mechanisms, and manifest as a continuum of changes as the infection progresses. Thus, signaling changes early in infection may induce pore or leak pathway changes. The durability and persistence of these responses may be dictated by pathogen burdens or specific virulence factors, as well as cross-talk between the pore and leak pathways. One example of cross-talk was demonstrated in the context of inflammatory bowel disease, where MLCK-dependent leak pathway permeability induced IL-13 expression; IL-13-dependent claudin-2 expression, in turn, selectively activated pore pathway permeability (145). MLCK-dependent leak pathway changes respond to rapid signaling changes, such as phosphorylation; since pore pathway changes require new protein synthesis, like claudin-2 expression, the responses manifest more gradually, but persist longer. Finally, with prolonged infection or high pathogen burdens, and attendant inflammatory infiltration, epithelial erosion could precipitate a complete collapse of barrier function (Fig. 5).
The consequences of barrier disruption include diarrhea due to the paracellular loss of water and ions, and increased penetration of microbes, toxins, and other molecules to the underlying tissues (42). The loss of TJ fence function can also result in the movement of basolateral proteins to apical sites, exposing new receptors for luminal organisms or toxins (83). In some instances, diarrhea can also be a host response that serves to reduce pathogen burdens.
The following sections discuss mechanisms by which a select group of microbes exploit TJs and alter paracellular permeability. Organisms belonging to the first three groups, namely attaching/effacing (A/E) pathogens, Salmonella enterica, and Shigella spp., are Gram-negative pathogens that use a syringe-like type III secretion system (T3SS) to alter host cell signaling. While the A/E pathogens are generally considered to be extracellular pathogens, Salmonella spp. and Shigella spp. occupy intracellular niches. Shigella spp. grows within the host cytoplasm, while Salmonella spp. resides within a specialized intracellular vacuolar compartment. The Gram-negative pathogen H. pylori is included for discussion since it colonizes and thrives in a distinct niche—the stomach. H. pylori uses a distinct secretion machinery, the type IV secretion system, to deliver a virulence protein into gastric epithelial cells. In the final section, three toxins produced by the Gram-positive clostridia are considered in the context of barrier disruption. Unlike the virulence proteins of the Gram-negative pathogens introduced above, the clostridial toxins are released into the environment and can often act as autonomous agents of disease. Notably, orally ingested botulinum toxin (BoNT) has features that allow it to navigate to peripheral neurons, with an early step involving intestinal barrier disruption.
Attaching and effacing pathogens
Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are diarrheagenic organisms that attach intimately to intestinal epithelial cells and efface the brush border microvilli (105). Related A/E pathogens Citrobacter rodentium and rabbit EPEC (REPEC) infect mice and rabbits, respectively, and are used to model human EPEC/EHEC infections (58). All A/E pathogens elaborate a T3SS, a bacterial membrane-associated syringe-like complex that delivers specific “effector” proteins directly into the host cytosol. The secreted molecules target numerous signaling pathways and alter host cell physiology, including TJ integrity and paracellular permeability (147).
The translocated intimin receptor (Tir) is a key A/E pathogen virulence protein secreted into host cells (147). Following entry, Tir inserts into the host cell membrane, and acts as a receptor for the bacterial surface adhesin, intimin. Tir-intimin interactions stimulate actin polymerization below the regions of bacterial attachment, eventually pushing the apical membrane up to form a pedestal-like structure.
There are three noteworthy features of EPEC secreted effector molecules which, incidentally, also hold true for several other pathogens. First, effector molecules are secreted into host cells in a hierarchical fashion, with Tir being the earliest protein delivered. Subsequently, other effectors are delivered in the following order: Tir < EspZ < NleA < EspF < EspH < Map < EspG (only a subset of effectors are indicated) (80). Second, each effector molecule may interact with several host proteins, and impact multiple functions. Finally, more than one effector may target a specific host cell function, such as paracellular permeability (136). The synergistic and complementary actions of effector molecules in mediating systematic temporal alterations in host cell physiology remains to be deciphered.
A/E pathogen-induced changes in TJ integrity and paracellular permeability
The first suggestion that A/E pathogens perturb TJs came from studies on rabbits infected with the rabbit diarrheagenic E. coli RDEC-1 (124). The investigators noted increased conductance across infected intestinal tissues, with no changes in the short-circuit current, and net equivalence of Na+ and Cl− transport. Subsequently, Canil et al. observed a drop in TER in EPEC-infected Caco-2 intestinal epithelial cells, but not in cells infected with adherence-defective mutants (13). The study, however, noted that [3H] inulin penetrance across the monolayer was not altered. Based on TER and sodium/mannitol flux studies in T84 intestinal epithelial monolayers, Spitz et al. concluded that EPEC infection disrupts TJs (119). These studies were subsequently confirmed, and correlated with the redistribution of ZO-1 and occludin from the TJs to intracellular compartments in infected T84 monolayers (24, 94, 111). Coimmunoprecipitation studies demonstrated decreased association between ZO-1, claudin, and occludin, and confocal microscopy revealed the progressive displacement of TJ proteins from apex to more basolateral locations of EPEC-infected cells (84). Freeze-fracture microscopy revealed the presence of claudin-1- and occludin-containing aberrant TJ strands along the lateral membrane of infected cells (84). In addition to barrier disruption, EPEC compromises the TJ fence function, which results in the movement of basolateral proteins to apical regions; this includes β1-integrin, which serves as an alternate/additional receptor for the EPEC adhesin, intimin (83).
In vivo A/E pathogen-induced barrier disruption was first confirmed in EPEC-infected C57BL/6J mice (107). Infected ileal and colonic tissues displayed significant decreases in TER, and these changes correlated with occludin redistribution to the cytoplasm. Other investigators replicated these observations, and also showed ZO-1 redistribution and increased biotin permeability in EPEC-infected mouse intestines (152). Similarly, the murine A/E pathogen C. rodentium also induced T3SS-dependent claudin-1, −3, and −5 displacement from the TJs following a 7-day infection period, and increased biotin permeability across infected monolayers, and into the lamina propria (43).
Virulence proteins involved in A/E pathogen-induced changes in TJ integrity and paracellular permeability
EPEC-induced TJ changes and barrier disruption require the T3SS-dependent delivery of effector proteins into intestinal epithelial cells. To date, five secreted molecules—EspF, Map, EspG, NleA, and EspH—have been implicated in infection-induced paracellular permeability changes (136). McNamara et al. first showed EspF to be essential for EPEC-induced occludin redistribution, TER decrease, and increased mannitol permeability in infected T84 cells (78). EspF was also required for the EPEC-induced displacement of occludin and claudin-1 from the TJs down into more lateral locations along the membrane (84). Dean et al. confirmed the requirement of EspF for EPEC-induced occludin redistribution and TER reduction in Caco-2 cells (25). Further, they showed the effector protein Map, and the adhesin intimin, to be independently required for inducing these changes.
EPEC strains lacking EspF (ΔespF) colonized C57BL/6J mice at levels comparable to the parent wildtype (WT) strain (107). Unlike WT EPEC, the ΔespF mutant failed to perturb occludin localization at the TJ, or reduce TER of the colon or ileum at 1 day postinfection. At 5 days postinfection, however, both the WT and ΔespF strains redistributed occludin and reduced TER, suggesting the involvement of other EPEC factors in mediating these changes. Similarly, unlike WT C. rodentium, an isogenic ΔespF mutant failed to redistribute claudin-3, or increase biotin permeabilization, in infected colonic tissue even in regions with robust colonization (43); a Δmap mutant, however, was similar to WT in altering claudin-3 distribution, ruling out a role for Map in this process. The investigators also found increased colonic luminal water content in C. rodentium WT (and Δmap)-infected animals relative to uninfected mice; ΔespF-infected animals showed an intermediate effect on luminal water content.
Role of EspF in paracellular permeability changes
EspF is a multifunctional protein that interacts with several host proteins, and has wide-ranging impacts on infected host cells (146). EspF localizes to the mitochondria and activates caspases; an L16E mutant was impaired for mitochondrial localization, but competent for barrier disruption (140). Independently, EspF interacts with sorting nexin 9 (SNX9), which regulates clathrin-mediated endocytosis (3). In a recent study, Tapia et al. demonstrated EspF-dependent redistribution of the Crumbs3 polarity complex, and the perturbation of epithelial cell polarity; site-directed EspF mutants deficient in SNX9 interaction failed to cause polarity changes (127). These mutants were not impaired for barrier disruption, suggesting independent effects of EspF on the polarity complex and on the TJ (3, 140).
EspF may regulate paracellular permeability of host cells by influencing the activation and localization of the membrane-cytoskeletal linker ezrin (110). Ezrin is phosphorylated and relocalized in LLC-PK1 (pig kidney cell line) cells infected with WT EPEC, but not a ΔespF mutant. Importantly, dominant-negative ezrin expression mitigated EPEC-induced ZO-1 relocalization, and TER decreases. The mechanisms by which EspF influences ezrin phosphorylation and increases its cytoskeletal association remain to be defined. Beyond ezrin, EspF may induce broader cytoskeletal alterations. EspF harbors neuronal Wiskott-Aldrich syndrome (N-WASP) binding motifs (2), which potentiate N-WASP/Arp2/3 actin nucleation. The N-WASP binding motifs were shown to be essential for EPEC EspF-induced epithelial barrier dysfunction (26). In studies with a rabbit strain of EPEC (E22), EspF was shown to bind N-WASP, Arp2/3, actin, and profilin, relocalize multiple TJ proteins to the pedestals, and trigger a reduction in TER (93).
Role of Map in paracellular permeability changes
The mechanisms by which Map influences paracellular permeability remain undefined. Map is essential for EPEC-induced TJ changes in Caco-2 cells, and constitutive Map expression in Madin-Darby Canine Kidney (MDCK) cells decreased TER, and increased the passage of both 4 kDa and 70 kDa dextran (112). On the other hand, a C. rodentium map deletion strain, like the parent WT strain, was able to alter barrier function of mouse colonic tissues (43). Since C. rodentium Δmap strains are impaired for causing diarrhea, and Map expression may be temporally regulated in vivo, more detailed studies are required to establish its role in paracellular permeability alterations (88, 127).
Role of NleA in paracellular permeability changes
Compared to WT EPEC, an NleA-deficient strain was impaired for decreasing the TER of T84 monolayers; plasmid-encoded NleA restored the phenotype to the ΔnleA strains (129). Occludin was lost from the periphery in EPEC-infected Caco-2 cells, but not in cells infected with ΔnleA, ΔespF, or Δmap strains, suggesting that all of these molecules are required to induce TJ alterations. NleA binds to, and inhibits, the COPII protein complex, which plays a role in trafficking intracellular proteins. Inhibition of vesicle trafficking by brefeldin A, a fungal compound, functionally restored the ability of ΔnleA (but not ΔespF or Δmap) strains to disrupt TJs (129). NleA-dependent TJ protein redistribution and altered paracellular permeability were confirmed in a C. rodentium/mouse model of infection. Thus, while EspF, and possibly Map, may directly disrupt TJs, NleA may potentiate this phenotype by blocking the trafficking of newly synthesized TJ proteins to the membrane.
Role of EspG1/2 in paracellular permeability changes
The related EPEC effectors EspG1 and EspG2 (42% identical, 62% similar) contribute to EPEC-induced TER decreases across epithelial cells (72,131). EspG disrupts host microtubule networks, and this promotes cytoplasmic accumulation of occludin and delays TJ recovery following infection (40).
Other host cell pathways involved in EPEC-mediated barrier disruption
Several studies have implicated specific host cell alterations in EPEC-induced barrier disruption, but the contributing effector molecules, and the proximal signaling pathways, have not been identified. Simonovic et al. (2000) demonstrated the dephosphorylation of occludin, and its displacement into intracellular compartments in EPEC-infected T84 cells (111). While the relevant occludin phosphorylation site(s) was not defined in this study, the serine/threonine phosphatase inhibitor calyculin A prevented EPEC-induced occludin relocalization, as well as infection-induced drop in TER. The atypical protein kinase C, PKCζ, has also been implicated in EPEC-induced TJ alterations and barrier disruption (130). PKCζ colocalized with occludin in infected cells, and PKCζ inhibition prevented EPEC-induced TER decrease. PKCζ is known to interact with the coiled-coil domain of occludin and regulate its phosphorylation and cellular localization (6, 90).
Role of actomyosin contraction in EPEC-induced barrier disruption
Early studies revealed MLC kinase to be one of the most prominent phosphoproteins in EPEC-infected cells (69). Subsequently, MLCK inhibition was shown to prevent EPEC-induced TER decrease in MDCK monolayers; a cell-permeable MLCK inhibitor also reduced infection-induced MLC phosphorylation, and attenuated EPEC-induced permeability changes, measured via TER and mannitol flux studies (150, 156). MLCK inhibition also ameliorated C. rodentium-induced colitis in mice (22).
It is worth highlighting that despite harboring closely related homologs and mediating similar gross alterations in host cell physiology, A/E pathogens exhibit differences at the mechanistic level. Thus, unlike EPEC, an EHEC ΔespF strain is not impaired for perturbing epithelial barrier function, measured as TER changes in infected T84 cells (139). EHEC espF, however, restored the ability of an EPEC ΔespF strain to decrease TER. Unlike EPEC, EHEC harbors EspF paralogs that may compensate for the absence of espF in the deletion strain. Also, unlike EPEC, EHEC infection had no impact on PKCζ expression or localization, and PKCζ inhibition did not impact EHEC-induced drop in TER in T84 cells (130).
Role of diarrhea in reducing pathogen burden
A recent study used a C. rodentium/mouse model of infection to demonstrate the beneficial effects of diarrhea for the host (132). C. rodentium infection resulted in IL-22-dependent upregulation of claudin-2, and increase in paracellular transport of water and Na+. Claudin-2-deficient mice had increased bacterial burdens, prolonged pathogen shedding, and greater tissue injury and disease; chemically-induced osmotic diarrhea reduced disease severity in these animals.
Salmonella enterica
Salmonella spp. are significant contributors to foodborne illness worldwide, with symptoms ranging from mild diarrhea to typhoid (57). These intracellular pathogens deploy two distinct T3SS, encoded on chromosomal regions called Salmonella pathogenicity islands (SPIs) 1 and 2, which inject virulence proteins into the host cytosol. SPI-1 T3SS effectors facilitate gut epithelial cell invasion, while SPI-2 effectors are essential for survival and growth of the bacteria within a specialized intracellular vacuolar compartment (57).
Salmonella-induced changes in TJ integrity and paracellular permeability
Finlay et al. first noted the reduction in TER in S. Cholerasuis-infected MDCK monolayers (30), and they subsequently confirmed these observations, and extended them to S. enterica serovar Typhimurium (S. Typhimurium) infections, in Caco-2 monolayers (29). Other investigators confirmed the paracellular permeability changes in Salmonella-infected MDCK II cells, including increased inulin transport (48). While contiguous ZO-1 membrane distribution was seen in infected cells, there were gross morphological TJ distortions, including contraction of the perijunctional actomyosin ring. A follow-up study using inhibitors, however, revealed that actomyosin contraction was not required for Salmonella-induced alterations in paracellular permeability (49). S. Typhimurium can modulate TJ integrity sufficiently to permit the migration of polymorphonuclear neutrophils across the intestinal monolayer (53).
Salmonella proteins that alter TJ integrity and paracellular permeability: SopB, SopE, SopE2, and SpiA
Initial clues to the potential mechanisms for Salmonella-induced barrier disruption came from Tafazoli et al., who observed that invasion-deficient mutants were unable to perturb ZO-1 or occludin distribution, increase paracellular dextran flux, or decrease TER (123). Further, the geranylger-anylltransferase 1 inhibitor GGTI-298, whose targets include Rho GTPases, blocked S. Typhimurium-induced TJ protein displacement, and TER decreases (123). Boyle et al. identified three SPI-1-encoded RhoGTPase-activating effectors, SopB, SopE, and SopE2, to be collectively required to mediate S. Typhimurium-dependent barrier alterations (11). SopB, SopE, and SopE2 induce other host cell alterations including the activation of a proinflammatory cascade that leads to neutrophil recruitment (57). While single mutations in the corresponding genes did not have a discernable impact (i.e., the mutants disrupted barrier function like the parent WT strain), a ΔsopB/E/E2 triple mutant failed to decrease TER or increase 4 kDa- and 40 kDa-dextran permeability. A ΔsipA/sopE/sopE2 strain was also severely impaired for disrupting barrier function. SipA is a SPI1-encoded effector that facilitates bacterial invasion via its actin bundling functions (57). The SPI-2 T3SS was dispensable for Salmonella-induced barrier disruption.
An earlier study presented data to suggest temporal shifts in Salmonella-induced TJ perturbations (7). Specifically, T84 cells infected with WT S. Typhimurium exhibited a rapid decrease in TER, while infection with a sigD-deficient mutant resulted in a delayed decrease in TER (SigD is also known as SopB). Both strains eventually lowered the TER to a similar extent. Bisindolylmaleimide, a conventional PKC inhibitor, inhibited the initial WT Salmonella-induced reduction in TER, but did not alter the eventual drop in resistance, suggesting a role for PKC in the acute SigD/SopB-dependent increase in paracellular permeability.
Zhang et al. observed a progressive increase in claudin-2 expression, as well as altered distribution of this protein at the TJs of S. Typhimurium-infected colonic epithelial cells (HT29C19A and SKCO15 cells), and in the colons of streptomycin-treated mice (154). siRNA-mediated claudin-2 knockdown attenuated infection-induced TER decrease, implicating the leak pathway in Salmonella-induced paracellular permeability changes. Using pharmacological inhibitors, the investigators implicated a role for the epidermal growth factor receptor pathway, and downstream c-Jun N-terminal kinases (JNK), in infection-induced claudin-2 expression. There was also a decrease in intracellular Salmonella in the siRNA-treated cells, implicating a direct or indirect role for claudin-2 in bacterial invasion.
A Salmonella protein that limits TJ disruption and paracellular permeability: AvrA
In contrast to the TJ-disrupting activities of SopB, SopE, SopE2, and SipA, another SPI-1 effector, AvrA, stabilizes TJs, and limits paracellular permeability (63). AvrA expression stabilized occludin and ZO-1 expression in S. Typhimurium-infected cells in vitro and in vivo, and transfected HT29C19A cells expressing AvrA had increased expression of occludin, ZO-1, and claudin-1. Compared to AvrA-deficient bacteria, strains harboring AvrA significantly decreased intestinal permeability to 3 kDa FITC-dextran in infected mice.
AvrA suppresses inflammation via its acetyltransferase activity toward specific host MAP kinases, and inhibition of the JNK/AP-1 and NF-κB signaling pathways (57). Extending the previous work, both S. Typhimurium and S. enterica Enteritidis (S. Enteritidis) AvrA were shown to augment TJ barrier function by stabilizing the expression and junctional localization of ZO-1 (64, 153). S. Enteritidis induced a dramatic decrease in TER, and this was further potentiated in ΔavrA-infected cells. Correspondingly, JNK phosphorylation was also greater in cells infected with the avrA deletion mutants. The JNK inhibitor SP600125 abolished AvrA-dependent ZO-1 stabilization at the TJs. Strains lacking avrA invaded epithelial cells better, and caused more pronounced pathology in a mouse gastroenteritis model of infection. AvrA also inhibited JNK pathway and stabilized ZO-1 levels in the mouse cecum. The two serovars, S. Typhimurium and S. Enteritidis did exhibit some differences; unlike S. Typhimurium, S. Enteritidis did not regulate occludin or claudin-1 and −2 levels.
It should be noted that WT S. Typhimurium prototype strain (14028s) expresses low levels of AvrA, and does not induce ZO-1 expression. The TJ-modulating functions of AvrA were uncovered using a 14028s derivative with an altered regulatory cascade that results in increased AvrA expression (among other alterations). The precise in vivo contexts in which the TJ modulatory functions of AvrA contribute to pathogenesis, therefore, remains to be defined. S. enterica strains associated with systemic infections have robust expression of SopB and SopE1, but do not express AvrA (63). One possible interpretation is that systemic Salmonella infection is facilitated by the ability to disrupt TJs.
Shigella spp.
Shigella spp. are the primary cause of bacillary dysentery, and the second leading cause (after rotavirus) of diarrheal deaths worldwide (20). Shigella is a primate-restricted pathogen, which distinguishes it from its close relatives. The four species of Shigella, S. dysenteriae, S. flexneri, S. boydii, and S. sonnei, vary in geographic distribution and antibiotic susceptibilities (73). These pathogens cross the epithelial monolayer via the M cells, access basolateral receptors of epithelial cells, and gain entry. They replicate within the host cytoplasm, and spread across to adjacent cells via a specialized mechanism (discussed below). A large virulence plasmid encodes most of the virulence factors including a T3SS.
Shigella-induced changes in TJ integrity and paracellular permeability
Sakaguchi et al. first explored the possibility that S. flexneri applied apically to polarized T84 cells could disrupt TJs and allow for the paracellular passage of bacteria (104). Apical infection with S. flexneri strains decreased the expression of ZO-1, ZO-2, and E-cadherin, dephosphorylated occludin, and displaced claudin-1 from membrane fractions of infected T84 cells. Both WT and noninvasive strains initially increased TER for 10 min postinfection, and subsequently reduced it to less than 70% of nonstimulated cells. Concomitantly, the bacteria penetrated the monolayer and gained access to the basolateral surface via the paracellular pathway. Apical infection of Caco-2 monolayers with S. flexneri or S. dysenteriae also caused the loss of ZO-1 from cell junctions and induced the hyperphosphorylation of occludin (32). These changes were accompanied by a decrease in TER, and increased paracellular permeability of 4 kDa dextran and bovine serum albumin (66 kDa).
Following entry and growth within the host cytosol, actin polymerization on one pole of the bacterium thrusts it forward, across the lateral membranes, and into adjacent cells. This allows the pathogen to spread across the epithelial monolayer without exposure to immune cells in the lumen or lamina propria. ZO-1 may be recruited to the distal portions of the Shigella actin tails, although the consequent implications to paracellular permeability is unknown (44). On reaching the inner surface of the host cell, the force of actin polymerization causes the bacteria to push against the membrane to form finger-like pseudopodia. Once inside the adjacent cell, the membranes are pinched off, and the bacteria are delivered into the new cell in a double-membraned compartment (4). The preferred points of cell-to-cell transit for the bacteria are junctions formed by three cells. The tricellular junction protein tricellulin is essential for cell-to-cell spread of Shigella, and involves a noncanonical clathrin-dependent endocytic pathway (35).
Shigella virulence factors that contribute to barrier disruption
The secreted protein SepA was recently implicated in Shigella-induced barrier disruption (68). SepA is the major S. flexneri protein secreted in culture and in infected cells, and is required for bacterial invasion of epithelial monolayers. WT S. flexneri, but not a sepA deletion strain (ΔsepA), dramatically reduced the expression of Lim kinase 1, an inhibitor of the actin-remodeling protein, cofilin. The consequent activation of cofilin triggers actin depolymerization and severing, leading to barrier disruption. SepA-mediated TJ changes facilitate bacterial penetration across the epithelium and subsequent bacterial invasion.
Helicobacter pylori
The gastric mucosal pathogen H. pylori colonizes over half of the human population and causes persistent gastritis in all carriers (95). Of the infected population, 10% to 15% develop severe gastric disease including adenocarcinoma. A unique set of virulence factors facilitate H. pylori to colonize its particularly hostile niche and cause disease. Some of these factors also alter TJs, a possible contributing event on the path to cancer development.
Helicobacter-induced changes in TJ integrity and paracellular permeability
Electron microscopy of biopsy samples from infected patients revealed the interaction of H. pylori (previously known as Campylobacter coli) with regions close to TJs, and the penetration of bacteria across the paracellular space (8, 17, 87). Freeze-fracture microscopy of antral (region close to the duodenum) biopsy samples from H. pylori-infected patients showed thickened, knobby, irregular, and focally fragmented TJ strands (89). H. pylori colonization was correlated with increased sucrose permeability, even in patients with asymptomatic gastritis (9, 34). Corpus biopsy samples from H. pylori-positive patients displayed greater paracellular permeability in Ussing chamber studies compared to tissues from H. pylori-negative individuals (74).
Helicobacter-induced alterations in permeability was recapitulated in animal models of infection. In Helicobacter felis-infected C57BL/6J mice, bacterial colonization and inflammation contributed to increased gastric permeability (75). Similarly, infection of C57BL/6J mice with the murine-adapted SS1 H. pylori strain resulted in altered distribution of cortical actin and occludin in the gastric surface epithelium. Lanthanum (4.2 A) tracer invasion assays confirmed barrier disruption of the gastric epithelium (122).
Mechanisms of H. pylori-induced alterations in paracellular permeability
In considering the impact of H. pylori on paracellular permeability, it is important to note that the structure and function of the gastric TJs are quite distinct from that of the intestine. Compared to the intestine, the stomach has a tighter epithelium, reflected in a greater baseline TER, and displays more TJ strands in freeze-fracture microscopy (14). Further, as in the intestine, there is heterogeneity in permeability in different parts of the gastric epithelium, with the surface epithelial cells being less permeable than those within the gastric glands (14). Some studies on the impact of H. pylori on paracellular permeability described below, particularly those using nongastric cells should, therefore, be interpreted with caution.
Most available gastric cell lines do not form monolayers and are, therefore, not useful for studying paracellular permeability changes. One exception is the NCI-N87 gastric cell line, which expresses claudin-18 (the most prominent isoform in gastric cells in vivo), forms a monolayer and has a baseline TER of about 1,000 Ω cm2. In gastric epithelial HGE-20 cells, which are derived from NCI-N87 cells, H. pylori induced the phosphorylation of IL-1 receptor, which then led to ROCK activation and displacement of claudin-4 from cell junctions to the cytosol (56). The consequences to paracellular permeability were not explored in this study. In the study by Fiorentino et al., H. pylori infection of NCI-N87 cells resulted in the loss of ZO-1 and claudin-1 from cell junctions, and caused a progressive decrease in TER relative to uninfected cells (31). H. pylori infection also increased the permeability to FITC-dextran (4 kDa) and FITC-BSA (40 kDa), and these changes were dependent on the presence of live bacteria.
Role of VacA in H. pylori-induced barrier disruption
The key virulence factors of H. pylori include the vacuolating toxin VacA, the cytotoxin-associated gene A (CagA), flagellin, and the enzyme urease (95). VacA was originally recognized for its ability to induce the formation of vacuolar compartments within host cells, but has subsequently been implicated in various other host cell phenotypic alterations. All H. pylori strains carry VacA, but there is considerable sequence variability across the group (95). Deletion of vacA had no impact on H. pylori-induced TER decreases in NCI-N87 gastric cells, suggesting that it does not play a role in increasing paracellular permeability (31). Similarly, VacA had no impact on paracellular permeability in Caco-2 and MKN28 gastric epithelial cells (149). In MDCK and T84 cells, however, purified and acid-activated VacA decreased TER, and increased mannitol and sucrose permeability, although there was no overt evidence of alterations in TJ protein distribution (91). In a follow-up study, the investigators reported that VacA-positive H. pylori, but not an isogenic ΔvacA strain, increased paracellular permeability (92).
Role of CagA in H. pylori-induced barrier disruption
The cag pathogenicity island is present in highly virulent strains and is a strong predictor for severe disease outcome, including gastric cancer (95). Genes within this island encode a type IV secretion system (T4SS, functionally akin to the T3SS discussed above, but composed of distinct components) which delivers effector proteins directly into the host cytosol. To date, CagA is the only known substrate secreted via the H. pylori T4SS.
As with VacA, evidence for a role for CagA in gastric epithelial paracellular perturbations is equivocal (14). CagA was not required for H. pylori-dependent paracellular permeability changes in Caco-2 intestinal epithelial cells, and MKN28 or HGE20 gastric epithelial cells (56, 65, 149). On the other hand, a cagA+ strain, but not an isogenic cagA− mutant, dramatically perturbed occludin and ZO-1 distribution in primary human gastric epithelial cells (55). Similarly, EM images revealed TJ alterations in AGS gastric epithelial cells infected with a cagA+ strain, but not a cagA− derivative (116). Neither of these studies, however, assessed paracellular permeability changes in the infected cells. In MDCK cells, injected CagA associated with ZO-1 and JAM, and induced the ectopic assembly of TJ complexes, and perturbed barrier function as assessed by ruthenium red staining and transmission electron microscopy (5). While CagA has been implicated in claudin gene regulation in some cell lines (claudin-2 in AGS cells, claudin-3 and −4 in T84 cells, and claudin-7 in MKN28 cells), the implications of these alterations to paracellular permeability is not known (28,116,148). Claudin isoforms are differentially altered in gastric cancer, and it is speculated that these changes play a role in tumorigenesis (47). CagA also perturbs epithelial cell polarity by interacting with components of the PAR polarity complex, leading to junctional defects and initiating epithelial-to-mesenchymal transition (128).
Role of urease in H. pylori-induced barrier disruption
H. pylori urease converts urea to ammonia (and carbon dioxide), which buffers gastric acid and creates a neutral zone around the bacterial surface (81). Urease-negative H. pylori mutants cannot colonize the stomach. Lytton and coworkers explored a potential role for the increased ammonium (NH4+) in gastric aspirates of H. pylori-infected patients to barrier disruption. Ammonium salt exposure, as well as WT H. pylori supernatants, acutely reduced the TER of Caco-2 cells, with a subsequent partial recovery. This was accompanied by TJ disruption and the expression of a low-molecular-weight form of occludin of 42 kDa. Epithelial cells exposed to supernatants of urease-deficient mutants did not exhibit a significant decrease in TER. Ammonia dampens the acid-dependent reinforcement of the epithelial barrier in gastric epithelial (HGE20) cells (70). Coculture of HE-20 cells with WT H. pylori, but not an isogenic urease-deficient strain (ΔureB), impeded acid-induced increase in TER.
Coculture of MKN28 gastric epithelial cells with WT H. pylori, but not an isogenic ureB mutant, resulted in a progressive decrease in TER (149). H. pylori increased MLC phosphorylation, which was blocked by inhibition of Rho kinase or MLCK; ureB-deficient bacteria failed to induce MLC phosphorylation. While Rho kinase can directly phosphorylate MLC, it can also inhibit MLC phosphatase activity by phosphorylating myosin phosphatase target subunit I (MYPT1). No significant changes were seen in phosphorylated MYPT1, suggesting a direct role for Rho kinase in phosphorylating MLC in H. pylori-infected cells.
Clostridial toxins and barrier function
The clostridia include numerous pathogenic strains capable of causing disease in humans and animals (96). These Gram-positive spore-forming organisms are the most prolific toxin producers among bacteria, and toxins are usually the main determinants of disease. Clostridial toxins belong to three broad groups: (a) pore-forming toxins like the cholesterol-dependent cytolysins, (b) nonpore-forming molecules, including enzymes, which act on host-cell surface molecules or matrix components, and (c) toxins that enter host cells and alter signaling pathways. Many of these toxins disrupt the intestinal epithelial barrier. In the following sections, one example from each of the above functional categories is discussed in the context of barrier disruption.
C. perfringens enterotoxin
C. perfringens enterotoxin (CPE) is responsible for the abdominal cramping and diarrhea associated with foodborne and non-foodborne enterotoxigenic C. perfringens strains (109). Ingested bacteria that escape killing by stomach acids multiply in the intestine and sporulate. CPE is released concomitant with spore egress from the mother cell. Katahira et al. first identified the CPE receptor, subsequently called as claudin-4, from an expression library of enterotoxin-sensitive Vero cells (51).
CPE-induced changes in TJ integrity and paracellular permeability
Sonoda et al. used CPE as a tool to probe the role of claudins in epithelial barrier function (117). The noncytotoxic C-terminal fragment of CPE (C-CPE) bound claudin-3 and −4 with high affinity, but not claudin-1 or −2, in MDCK cells. Basolateral, but not apical, C-CPE treatment of MDCK1 caused the selective displacement of claudin-4 from the TJs to the cytoplasm, and its subsequent degradation. Freeze-fracture microscopy showed aberrant TJs, with reduced number of strands and limited network complexity. Basolateral treatment of MDCK cells with C-CPE induced a ~4.5-fold drop in TER, and a twofold increase in the flux of 4 kDA and 10 kDA FITC-dextran, but not 40 kDa FITC-dextran. Removal of C-CPE resulted in the gradual recovery of TER to the levels of untreated controls. This was, in fact, the first evidence of a role for claudins in epithelial barrier function.
Members of the claudin family serve as high (claudin-3, −4, −6, −7, and 9) or low/medium (claudin-1, 2, 8, 14, and 19) affinity receptors for CPE (27). CPE is an unusual pore-forming toxin that recruits host membrane proteins, including occludin, into large membrane complexes of up to 200 kDa (114). Complex formation was shown to be essential for CPE cytotoxicity. At low concentrations (1 μg/mL), CPE forms a limited number of pores that allows passage of small cations, including the influx of Ca2+, which leads to rapid activation of the intrinsic apoptosis pathway (15). In Caco-2 cells, 1 μg/mL CPE disrupted the actin cytoskeleton and induced the removal of occludin, but not ZO-1, from the TJs (113).
While the C-CPE fragment perturbed TJs and altered barrier function without a cytotoxic effect on host cells (117), this pathway may not be involved in the permeability increases mediated by the intact toxin. Based on the kinetics of killing, it has been argued that CPE perturbs barrier function primarily via its pore-forming and cytotoxic effects (27).
Clostridium difficile toxins A and B
Clostridium difficile, the leading cause of healthcare-associated diarrhea, was responsible for nearly half a million cases in 2011 in the United States, with 29,300 deaths (62). Ingested spores germinate into vegetative cells in the intestine in response to certain bile acids and glycine. Antibiotic-mediated alterations of the normal flora allows the vegetative cells to colonize and produce toxins. The large clostridial toxins, toxin A (TcdA) and toxin B (TcdB), are the primary mediators of disease; the binary toxin (CDT), present in some strains, is associated with increased patient mortality (38).
While TcdA and TcdB share sequence similarity, they bind to distinct receptors and have unique effects on host cells, and play different roles in vivo (115). Both toxins have a C-terminal combined repetitive oligopeptide repeat (CROP) domain that facilitates interactions with host cell oligosaccharides and facilitates toxin endocytosis. TcdA binds to multiple glycolipids and glycosylated proteins, while TcdB receptors include the poliovirus receptor-like protein (PVRL3, also known as nectin-3) and chondroitin sulfate proteoglycan 4. PVRL3/nectin-3 is a component of AJs which interacts with afadin, a filamentous actin-binding protein. Following entry and endosome acidification, the central cysteine protease domain and delivery domains facilitate autocatalytic processing of the toxin and egress of the N-terminal glucosyl transferase domain (GTD) into the cytosol. The GTD modifies and inactivates Rho family GTPases including Rho, Rac, and CDC42, resulting in cytoskeletal alterations and host cell death.
Rodent studies with isogenic TcdA or TcdB mutants concur on the ability of TcdB to cause severe disease in the absence of TcdA. Consistent with this, Tcd A−B+ strains have been recovered in clinical settings (2). On the other hand, data on the ability of Tcd A+B− strains to cause disease have been more variable (115). In ileal loop models, TcdA caused profound necrosis and inflammation, while even high levels of TcdB failed to induce these changes. In studies with colonic tissues, TcdB is more proinflammatory than TcdA.
C. difficile toxin-induced changes in TJ integrity and paracellular permeability
Hecht and coworkers first demonstrated the disruption of barrier function by C. difficile TcdA and TcdB (45, 46), and Moore et al. confirmed TcdA-dependent permeability changes in ex vivo experiments using guinea pig ileal tissues (82). The addition of either toxin to the apical surface of T84 cells increased paracellular permeability, as measured via TER and sodium-mannitol flux studies, but TcdA-induced effects were markedly more potent and rapid (TER decreases of ~99% in 6 to 8 h for TcdA, ~74% in 72 h for TcdB, both toxins at 0.7 μg/mL) (45, 46). While the toxins caused the disassembly of perijunctional actin filaments, TcdA-dependent TJ permeability increases preceded these changes. Chen et al. explored the possibility that TcdA-mediated barrier disruption occurred prior to, and independent of Rho glucosylation (16). They demonstrated the TcdA-dependent activation of PKCα and PKCβ, increased ZO-1 translocation from TJs, and decrease in TER of T84 cells within 2 to 3 h of toxin exposure. Consistent with a role for TcdA-induced PKC activation in barrier disruption, a PKCα/β antagonist inhibited TcdA-mediated TJ protein changes and TER decrease. Other investigators have also reported the glucosyltransferase-independent impacts of the toxins on epithelial barrier function (151).
The reasons underlying the delayed TER decrease for TcdB relative to TcdA is presently not known (45, 46). TcdB, unlike TcdA, may impact barrier function only after entry into cells and subsequent Rho GTPase inhibition. It is also possible that TcdB interferes with cell-cell interactions by binding to nectin-3 and disrupting the AJs. It should be noted, however, that nectin-3 knockout mice did not show significant alterations in paracellular permeability (125).
Based on the differential impacts of the two toxins when added to the apical surface of epithelial cells, a synergistic mode of action has been proposed (102). According to this model, TcdA-mediated barrier disruption results in the increased paracellular passage of TcdB. Interaction of TcdB with basolateral receptors, possibly PVRL3/nectin-3, then accounts for the marked increase in cytotoxic effects and further damage to the epithelium. A caveat to this model is that Tcd A−B+ C. difficile strains have been associated with outbreaks, and can cause disease in animal models (2, 10). Supernatants from two outbreak-associated Tcd A−B+ strains failed to decrease the TER of Caco-2 cells even after 10 h incubation, while a Tcd A+B+ strain dramatically reduced TER within 5 h (2). Thus, TcdA may facilitate TcdB translocation and intoxication, but there are likely independent pathways for TcdB to breach the epithelial barrier and cause disease.
Botulinum toxin
One of the most potent toxins known, BoNTs are produced by Clostridium botulinum, and cause the persistent paralysis of peripheral nerve terminals, resulting in the disease known as botulism (100). In foodborne and infant botulism, the most common forms, the ingested toxin must cross the intestinal epithelial barrier, enter the systemic circulation, and eventually reach target neurons. BoNTs proteolytically cleave peripheral nerve SNARE proteins and block neurotransmitter release. To date, more than 40 genetic variants of BoNT, differing up to 36% at the amino acid level, have been identified (101). BoNTs were traditionally classified on the basis of interactions with monovalent botulinum antitoxin into seven serotypes (A-G), but this system has come into question with the identification of newer cross-reacting BoNT types. BoNT types A, B, E, and F cause botulism in humans, while types C and D cause disease in birds and cattle, respectively.
BoNTs form large protein complexes by associating with neurotoxin-associated proteins (NAPs) (100). NAPs include nontoxic nonhemagglutinin (NTNHA) and hemagglutinin (HA), which itself is comprised of three proteins, HA1, HA2, and HA3. The large progenitor toxin complex (L-PTC) or 16S complex, which contains BoNT, NTNHA, and HA, has an oral toxicity that is several hundred folds more than that of BoNT itself (71). Nontoxic components protect BoNT from proteases and other damaging agents in the gastrointestinal tract. In addition, the HA complex facilitates toxin transport across the intestinal epithelium.
BoNT-induced changes in TJ integrity and paracellular permeability
Matsumura and coworkers showed that L-PTC added to either side of Caco-2 monolayers induced a drop in TER; the effect was more pronounced and rapid when the toxin was added basolaterally. In MDCK cells, however, L-PTC decreased the TER only when added basolaterally. In contrast, neither the M-PTC (BoNT + NTNHA) nor BoNT alone decreased the TER of Caco-2 cells, suggesting a role for the HA complex in this process. Further, L-PTC-induced paracellular permeability changes were blocked by anti-HA antiserum. Apical addition of L-PTC also disrupted the localization of occludin, ZO-1, E-cadherin, and β-catenin at cell boundaries, and increased the paracellular flux of dextran (4–150 kDa). The permeability changes were not accompanied by any toxic effects on the Caco-2 cells.
In subsequent work, this group showed the direct interaction of HA with the human, bovine and mouse isoforms of E-cadherin, but not those of rats or chickens (121). Recombinant HA caused E-cadherin internalization and disrupted cell-cell adhesion. HA also decreased the TER of MDCK cells expressing mouse, but not rat, E-cadherin. L-PTC/A and L-PTC/B, but not L-PTC/C, interacted with E-cadherin (121); correspondingly, L-PTC/A and L-PTC/B, but not L-PTC/C, decreased TER and increased 4 to 150 kDa dextran flux across Caco-2 and T84 monolayer (50).
E-cadherin, a component of the AJs, is present below the TJs, and would generally not be accessible to HA from the apical surface. This poses a conundrum: for HA to interact with E-cadherin and disrupt the barrier, it would first have to access the basolateral surface. Available data support two modes by which HA can reach the basal side: transcytosis through epithelial cells, or via M cells. HA added to the apical surface of Caco-2 cells was internalized into endosomes and transcytosed to the basolateral sides (121). HA also bound to glycoprotein 2 on microfold (M) cells in the Peyer’s patches of mouse intestines, and facilitated transport of the toxin complex to the basolateral sides (71). PTC lacking HA (M-PTC) did not localize to Peyer’s patches, and were two orders of magnitude less toxic when delivered orally to mice. Mice depleted for M cells, or lacking glycoprotein 2, were markedly less susceptible to orally delivered L-PTC/A. Traversing through the M cells, the toxin complex is delivered to the basolateral side.
If transcytosis can deliver the toxin to the basolateral side, is there a role for intestinal epithelial barrier disruption in the intoxication process? Lee and coworkers generated HA complexes with single-amino acid changes in two of the HA proteins which were deficient for E-cadherin binding, but not for transcytosis across epithelial monolayers (61). This complex failed to increase paracellular permeability. L-PTC/A complexes with the mutant HA proteins were severely impaired for intoxication in a mouse model, suggesting HA-dependent junctional perturbations substantially augment BoNT transport via the paracellular pathway to the target neurons. Finally, type E and type F toxins produce toxin complexes that lack HA, but still cause food-borne botulism, though they have less oral toxicity (103). Thus, the HA proteins, while not essential, can increase the oral toxicity of BoNT.
Conclusion
The maintenance of intestinal epithelial barrier function is critical for health. In the examples cited above, TJ disruption and altered barrier function not only contribute to disease symptoms but often represent a transition phase in the pathogen’s virulence program. Thus, increased permeability may allow pathogens or toxins to penetrate deeper tissue, while increased luminal nutrients and water could promote pathogen growth and dispersal. Recent studies also suggest a host-protective value for increased paracellular permeability and diarrhea. Our understanding of the dynamics of the junctional barrier has been expanding rapidly over the past few years. Studies on pathogen-induced barrier disruption will have to be viewed fresh in light of the recent findings. A detailed understanding of the mechanistic and temporal parameters of TJ alterations during infections could offer novel opportunities for mitigating disease.
Didactic Synopsis.
Major teaching points
Paracellular transport is the passive movement of ions, solutes, and water via the spaces between cells. This process is regulated by the tight junction complex.
There are two recognized modes of paracellular transport—the pore and leak pathways—that are independently, and sometimes opposingly, regulated.
The high-capacity pore pathway is ion- and size-selective, and is gated by the claudin family of proteins. The low-capacity, charge-insensitive, leak pathway permits passage of larger molecules.
Many pathogenic microbes and their products target tight junctions, and alter their composition and function. The consequent perturbation of paracellular permeability contributes to disease symptoms, including diarrhea.
Concomitant with altered permeability, the loss of epithelial cell polarity, and the potential passage of toxins and microbes into the lamina propria may exacerbate disease.
Commensal and probiotic microbes and their products can mitigate pathogen-induced perturbations of paracellular permeability, although the molecular mechanisms are not always well defined.
Acknowledgements
The Viswanathan laboratory is supported by the National Institutes of Health (V. K. NIAID1R01AI081742) and the USDA CSREES Hatch Program (ARZT-5704100-A02-140). We are grateful to Dr. Gayatri Vedantam for critically reading the manuscript and suggesting substantial improvements. We thank Drs. Yong-Guo Zhang and Jun Sun, University of Illinois at Chicago, Chicago, IL, for kindly sharing unpublished images showing claudin expression changes in Salmonella-infected intestinal epithelial cells.
References
- 1.Al-Sadi R, Khatib K, Guo S, Ye D, Youssef M, Ma T. Occludin regulates macromolecule flux across the intestinal epithelial tight junction barrier. Am J Physiol Gastrointest Liver Physiol 300: G1054–G1064, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Alfa MJ, Kabani A, Lyerly D, Moncrief S, Neville LM, Al-Barrak A, Harding GK, Dyck B, Olekson K, Embil JM. Characterization of a toxin A-negative, toxin B-positive strain of Clostridium difficile responsible for a nosocomial outbreak of Clostridium difficile-associated diarrhea. J Clin Microbiol 38: 2706–2714, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Alto NM, Weflen AW, Rardin MJ, Yarar D, Lazar CS, Tonikian R, Koller A, Taylor SS, Boone C, Sidhu SS, Schmid SL, Hecht GA, Dixon JE. The type III effector EspF coordinates membrane trafficking by the spatiotemporal activation of two eukaryotic signaling pathways. J Cell Biol 178: 1265–1278, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Amieva M Shigella navigates tight corners. Cell Host Microbe 11: 319–320, 2012. [DOI] [PubMed] [Google Scholar]
- 5.Amieva MR, Vogelmann R, Covacci A, Tompkins LS, Nelson WJ, Falkow S. Disruption of the epithelial apical-junctional complex by Helicobacter pylori CagA. Science 300: 1430–1434, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Andreeva AY, Krause E, Muller EC, Blasig IE, Utepbergenov DI. Protein kinase C regulates the phosphorylation and cellular localization of occludin. J Biol Chem 276: 38480–38486, 2001. [DOI] [PubMed] [Google Scholar]
- 7.Bertelsen LS, Paesold G, Marcus SL, Finlay BB, Eckmann L, Barrett KE. Modulation of chloride secretory responses and barrier function of intestinal epithelial cells by the Salmonella effector protein SigD. Am J Physiol Cell Physiol 287: C939–C948, 2004. [DOI] [PubMed] [Google Scholar]
- 8.Bode G, Malfertheiner P, Ditschuneit H. Pathogenetic implications of ultrastructural findings in Campylobacter pylori related gastroduodenal disease. Scand J Gastroenterol Suppl 142: 25–39, 1988. [PubMed] [Google Scholar]
- 9.Borch K, Sjostedt C, Hannestad U, Soderholm JD, Franzen L, Mardh S. Asymptomatic Helicobacter pylori gastritis is associated with increased sucrose permeability. Dig Dis Sci 43: 749–753, 1998. [DOI] [PubMed] [Google Scholar]
- 10.Borriello SP, Wren BW, Hyde S, Seddon SV, Sibbons P, Krishna MM, Tabaqchali S, Manek S, Price AB. Molecular, immunological, and biological characterization of a toxin A-negative, toxin B-positive strain of Clostridium difficile. Infect Immun 60: 4192–4199, 1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Boyle EC, Brown NF, Finlay BB. Salmonella enterica serovar Typhimurium effectors SopB, SopE, SopE2 and SipA disrupt tight junction structure and function. Cell Microbiol 8: 1946–1957, 2006. [DOI] [PubMed] [Google Scholar]
- 12.Buschmann MM, Shen L, Rajapakse H, Raleigh DR, Wang Y, Wang Y, Lingaraju A, Zha J, Abbott E, McAuley EM, Breskin LA, Wu L, Anderson K, Turner JR, Weber CR. Occludin OCEL-domain interactions are required for maintenance and regulation of the tight junction barrier to macromolecular flux. Mol Biol Cell 24: 3056–3068, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Canil C, Rosenshine I, Ruschkowski S, Donnenberg MS, Kaper JB, Finlay BB. Enteropathogenic Escherichia coli decreases the transepithelial electrical resistance of polarized epithelial monolayers. Infect Immun 61: 2755–2762, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Caron TJ, Scott KE, Fox JG, Hagen SJ. Tight junction disruption: Helicobacter pylori and dysregulation of the gastric mucosal barrier. World J Gastroenterol 21: 11411–11427, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chakrabarti G, Zhou X, McClane BA. Death pathways activated in CaCo-2 cells by Clostridium perfringens enterotoxin. Infect Immun 71: 4260–4270, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chen ML, Pothoulakis C, LaMont JT. Protein kinase C signaling regulates ZO-1 translocation and increased paracellular flux of T84 colonocytes exposed to Clostridium difficile toxin A. J Biol Chem 277: 4247–4254, 2002. [DOI] [PubMed] [Google Scholar]
- 17.Chen XG, Correa P, Offerhaus J, Rodriguez E, Janney F, Hoffmann E, Fox J, Hunter F, Diavolitsis S. Ultrastructure of the gastric mucosa harboring Campylobacter-like organisms. Am J Clin Pathol 86: 575–582, 1986. [DOI] [PubMed] [Google Scholar]
- 18.Citalan-Madrid AF, Vargas-Robles H, Garcia-Ponce A, Shibayama M, Betanzos A, Nava P, Salinas-Lara C, Rottner K, Mennigen R, Schnoor M. Cortactin deficiency causes increased RhoA/ROCK1-dependent actomyosin contractility, intestinal epithelial barrier dysfunction, and disproportionately severe DSS-induced colitis. Mucosal Immunol 10: 1237–1247, 2017. [DOI] [PubMed] [Google Scholar]
- 19.Claude P, Goodenough DA. Fracture faces of zonulae occludentes from “tight” and “leaky” epithelia. J Cell Biol 58: 390–400, 1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Collaborators GBDDD. Estimates of global, regional, and national morbidity, mortality, and aetiologies of diarrhoeal diseases: A systematic analysis for the Global Burden of Disease Study 2015. Lancet Infect Dis 17(9): 909–948, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Colpitts CC, Baumert TF. Claudins in viral infection: From entry to spread. Pflugers Arch 469: 27–34, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Conlin VS, Wu X, Nguyen C, Dai C, Vallance BA, Buchan AM, Boyer L, Jacobson K. Vasoactive intestinal peptide ameliorates intestinal barrier disruption associated with Citrobacter rodentium-induced colitis. Am J Physiol Gastrointest Liver Physiol 297: G735–G750, 2009. [DOI] [PubMed] [Google Scholar]
- 23.Cunningham KE, Turner JR. Myosin light chain kinase: Pulling the strings of epithelial tight junction function. Ann N Y Acad Sci 1258: 34–42, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Czerucka D, Dahan S, Mograbi B, Rossi B, Rampal P. Saccharomyces boulardii preserves the barrier function and modulates the signal transduction pathway induced in enteropathogenic Escherichia coli-infected T84 cells. Infect Immun 68: 5998–6004, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dean P, Kenny B. Intestinal barrier dysfunction by enteropathogenic Escherichia coli is mediated by two effector molecules and a bacterial surface protein. Mol Microbiol 54: 665–675, 2004. [DOI] [PubMed] [Google Scholar]
- 26.Dean P, Young L, Quitard S, Kenny B. Insights into the pathogenesis of enteropathogenic E. coli using an improved intestinal enterocyte model. PLoS One 8: e55284, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Eichner M, Protze J, Piontek A, Krause G, Piontek J. Targeting and alteration of tight junctions by bacteria and their virulence factors such as Clostridium perfringens enterotoxin. Pflugers Arch 469: 77–90, 2017. [DOI] [PubMed] [Google Scholar]
- 28.El-Etr SH, Mueller A, Tompkins LS, Falkow S, Merrell DS. Phosphorylation-independent effects of CagA during interaction between Helicobacter pylori and T84 polarized monolayers. J Infect Dis 190: 1516–1523, 2004. [DOI] [PubMed] [Google Scholar]
- 29.Finlay BB, Falkow S. Salmonella interactions with polarized human intestinal Caco-2 epithelial cells. J Infect Dis 162: 1096–1106, 1990. [DOI] [PubMed] [Google Scholar]
- 30.Finlay BB, Gumbiner B, Falkow S. Penetration of Salmonella through a polarized Madin-Darby canine kidney epithelial cell monolayer. J Cell Biol 107: 221–230, 1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fiorentino M, Ding H, Blanchard TG, Czinn SJ, Sztein MB, Fasano A. Helicobacter pylori-induced disruption of monolayer permeability and proinflammatory cytokine secretion in polarized human gastric epithelial cells. Infect Immun 81: 876–883, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fiorentino M, Levine MM, Sztein MB, Fasano A. Effect of wild-type Shigella species and attenuated Shigella vaccine candidates on small intestinal barrier function, antigen trafficking, and cytokine release. PLoS One 9: e85211, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.France MM, Turner JR. The mucosal barrier at a glance. J Cell Sci 130: 307–314, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fukuda Y, Bamba H, Okui M, Tamura K, Tanida N, Satomi M, Shimoyama T, Nishigami T. Helicobacter pylori infection increases mucosal permeability of the stomach and intestine. Digestion 63 (Suppl 1): 93–96, 2001. [DOI] [PubMed] [Google Scholar]
- 35.Fukumatsu M, Ogawa M, Arakawa S, Suzuki M, Nakayama K, Shimizu S, Kim M, Mimuro H, Sasakawa C. Shigella targets epithelial tricellular junctions and uses a noncanonical clathrin-dependent endocytic pathway to spread between cells. Cell Host Microbe 11: 325–336, 2012. [DOI] [PubMed] [Google Scholar]
- 36.Furuse M, Furuse K, Sasaki H, Tsukita S. Conversion of zonulae occludentes from tight to leaky strand type by introducing claudin-2 into Madin-Darby canine kidney I cells. J Cell Biol 153: 263–272, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Garcia-Hernandez V, Quiros M, Nusrat A. Intestinal epithelial claudins: Expression and regulation in homeostasis and inflammation. Ann N Y Acad Sci 1397: 66–79, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gerding DN, Johnson S, Rupnik M, Aktories K. Clostridium difficile binary toxin CDT: Mechanism, epidemiology, and potential clinical importance. Gut Microbes 5: 15–27, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ghandehari H, Smith PL, Ellens H, Yeh PY, Kopecek J. Size-dependent permeability of hydrophilic probes across rabbit colonic epithelium. J Pharmacol Exp Ther 280: 747–753, 1997. [PubMed] [Google Scholar]
- 40.Glotfelty LG, Zahs A, Hodges K, Shan K, Alto NM, Hecht GA. Enteropathogenic E. coli effectors EspG1/G2 disrupt microtubules, contribute to tight junction perturbation and inhibit restoration. Cell Microbiol 16: 1767–1783, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gonzalez-Mariscal L, Chavez de Ramirez B, Lazaro A, Cereijido M. Establishment of tight junctions between cells from different animal species and different sealing capacities. J Membr Biol 107: 43–56, 1989. [DOI] [PubMed] [Google Scholar]
- 42.Guttman JA, Finlay BB. Tight junctions as targets of infectious agents. Biochim Biophys Acta 1788: 832–841, 2009. [DOI] [PubMed] [Google Scholar]
- 43.Guttman JA, Li Y, Wickham ME, Deng W, Vogl AW, Finlay BB. Attaching and effacing pathogen-induced tight junction disruption in vivo. Cell Microbiol 8: 634–645, 2006. [DOI] [PubMed] [Google Scholar]
- 44.Hanajima-Ozawa M, Matsuzawa T, Fukui A, Kamitani S, Ohnishi H, Abe A, Horiguchi Y, Miyake M. Enteropathogenic Escherichia coli, Shigella flexneri, and Listeria monocytogenes recruit a junctional protein, zonula occludens-1, to actin tails and pedestals. Infect Immun 75: 565–573, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hecht G, Koutsouris A, Pothoulakis C, LaMont JT, Madara JL. Clostridium difficile toxin B disrupts the barrier function of T84 monolayers. Gastroenterology 102: 416–423, 1992. [DOI] [PubMed] [Google Scholar]
- 46.Hecht G, Pothoulakis C, LaMont JT, Madara JL. Clostridium difficile toxin A perturbs cytoskeletal structure and tight junction permeability of cultured human intestinal epithelial monolayers. J Clin Invest 82: 1516–1524, 1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Iravani O, Tay BW, Chua PJ, Yip GW, Bay BH. Claudins and gastric carcinogenesis. Exp Biol Med (Maywood) 238: 344–349, 2013. [DOI] [PubMed] [Google Scholar]
- 48.Jepson MA, Collares-Buzato CB, Clark MA, Hirst BH, Simmons NL. Rapid disruption of epithelial barrier function by Salmonella Typhimurium is associated with structural modification of intercellular junctions. Infect Immun 63: 356–359, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jepson MA, Schlecht HB, Collares-Buzato CB. Localization of dysfunctional tight junctions in Salmonella enterica serovar Typhimurium-infected epithelial layers. Infect Immun 68: 7202–7208, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jin Y, Takegahara Y, Sugawara Y, Matsumura T, Fujinaga Y. Disruption of the epithelial barrier by botulinum haemagglutinin (HA) proteins—Differences in cell tropism and the mechanism of action between HA proteins of types A or B, and HA proteins of type C. Microbiology 155: 35–45, 2009. [DOI] [PubMed] [Google Scholar]
- 51.Katahira J, Inoue N, Horiguchi Y, Matsuda M, Sugimoto N. Molecular cloning and functional characterization of the receptor for Clostridium perfringens enterotoxin. J Cell Biol 136: 1239–1247, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kim S, Covington A, Pamer EG. The intestinal microbiota: Antibiotics, colonization resistance, and enteric pathogens. Immunol Rev 279: 90–105, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kohler H, Sakaguchi T, Hurley BP, Kase BA, Reinecker HC, McCormick BA. Salmonella enterica serovar Typhimurium regulates intercellular junction proteins and facilitates transepithelial neutrophil and bacterial passage. Am J Physiol Gastrointest Liver Physiol 293: G178–G187, 2007. [DOI] [PubMed] [Google Scholar]
- 54.Krug SM, Gunzel D, Conrad MP, Rosenthal R, Fromm A, Amasheh S, Schulzke JD, Fromm M. Claudin-17 forms tight junction channels with distinct anion selectivity. Cell Mol Life Sci 69: 2765–2778, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lai YP, Yang JC, Lin TZ, Lin JT, Wang JT. Helicobacter pylori infection and CagA protein translocation in human primary gastric epithelial cell culture. Helicobacter 11: 451–459, 2006. [DOI] [PubMed] [Google Scholar]
- 56.Lapointe TK, O’Connor PM, Jones NL, Menard D, Buret AG. Interleukin-1 receptor phosphorylation activates Rho kinase to disrupt human gastric tight junctional claudin-4 during Helicobacter pylori infection. Cell Microbiol 12: 692–703, 2010. [DOI] [PubMed] [Google Scholar]
- 57.LaRock DL, Chaudhary A, Miller SI. Salmonellae interactions with host processes. Nat Rev Microbiol 13: 191–205, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Law RJ, Gur-Arie L, Rosenshine I, Finlay BB. In vitro and in vivo model systems for studying enteropathogenic Escherichia coli infections. Cold Spring Harb Perspect Med 3: a009977, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lechuga S, Baranwal S, Ivanov AI. Actin-interacting protein 1 controls assembly and permeability of intestinal epithelial apical junctions. Am J Physiol Gastrointest Liver Physiol 308: G745–G756, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lechuga S, Ivanov AI. Disruption of the epithelial barrier during intestinal inflammation: Quest for new molecules and mechanisms. Biochim Biophys Acta 1864: 1183–1194, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lee K, Zhong X, Gu S, Kruel AM, Dorner MB, Perry K, Rummel A, Dong M, Jin R. Molecular basis for disruption of E-cadherin adhesion by botulinum neurotoxin A complex. Science 344: 1405–1410, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, Farley MM, Holzbauer SM, Meek JI, Phipps EC, Wilson LE, Winston LG, Cohen JA, Limbago BM, Fridkin SK, Gerding DN, McDonald LC. Burden of Clostridium difficile infection in the United States. N Engl J Med 372: 825–834, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liao AP, Petrof EO, Kuppireddi S, Zhao Y, Xia Y, Claud EC, Sun J. Salmonella type III effector AvrA stabilizes cell tight junctions to inhibit inflammation in intestinal epithelial cells. PLoS One 3: e2369, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lin Z, Zhang YG, Xia Y, Xu X, Jiao X, Sun J. Salmonella enteritidis effector AvrA stabilizes intestinal tight junctions via the JNK pathway. J Biol Chem 291: 26837–26849, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lytton SD, Fischer W, Nagel W, Haas R, Beck FX. Production of ammonium by Helicobacter pylori mediates occludin processing and disruption of tight junctions in Caco-2 cells. Microbiology 151: 3267–3276, 2005. [DOI] [PubMed] [Google Scholar]
- 66.Ma TY, Iwamoto GK, Hoa NT, Akotia V, Pedram A, Boivin MA, Said HM. TNF-α-induced increase in intestinal epithelial tight junction permeability requires NF-κB activation. Am J Physiol Gastrointest Liver Physiol 286: G367–G376, 2004. [DOI] [PubMed] [Google Scholar]
- 67.Madara JL. Functional morphology of epithelium of the small intestine. In: Comprehensive Physiology 83–120, 2010. [Google Scholar]
- 68.Maldonado-Contreras A, Birtley JR, Boll E, Zhao Y, Mumy KL, Toscano J, Ayehunie S, Reinecker HC, Stern LJ, McCormick BA. Shigella depends on SepA to destabilize the intestinal epithelial integrity via cofilin activation. Gut Microbes 8(6): 544–560, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Manjarrez-Hernandez HA, Baldwin TJ, Williams PH, Haigh R, Knutton S, Aitken A. Phosphorylation of myosin light chain at distinct sites and its association with the cytoskeleton during enteropathogenic Escherichia coli infection. Infect Immun 64: 2368–2370, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Marcus EA, Vagin O, Tokhtaeva E, Sachs G, Scott DR. Helicobacter pylori impedes acid-induced tightening of gastric epithelial junctions. Am J Physiol Gastrointest Liver Physiol 305: G731–G739, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Matsumura T, Sugawara Y, Yutani M, Amatsu S, Yagita H, Kohda T, Fukuoka S, Nakamura Y, Fukuda S, Hase K, Ohno H, Fujinaga Y. Botulinum toxin A complex exploits intestinal M cells to enter the host and exert neurotoxicity. Nat Commun 6: 6255, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Matsuzawa T, Kuwae A, Abe A. Enteropathogenic Escherichia coli type III effectors EspG and EspG2 alter epithelial paracellular permeability. Infect Immun 73: 6283–6289, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mattock E, Blocker AJ. How do the virulence factors of Shigella work together to cause disease?. Front Cell Infect Microbiol 7: 64, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Matysiak-Budnik T, Coffin B, Lavergne-Slove A, Sabate JM, Megraud F, Heyman M. Helicobacter pylori increases the epithelial permeability to a food antigen in human gastric biopsies. Am J Gastroenterol 99: 225–232, 2004. [DOI] [PubMed] [Google Scholar]
- 75.Matysiak-Budnik T, Hashimoto K, Heyman M, de Mascarel A, Desjeux JF, Megraud F. Antral gastric permeability to antigens in mice is altered by infection with Helicobacter felis. Eur J Gastroenterol Hepatol 11: 1371–1377, 1999. [DOI] [PubMed] [Google Scholar]
- 76.McKee ML, O’Brien AD. Investigation of enterohemorrhagic Escherichia coli O157:H7 adherence characteristics and invasion potential reveals a new attachment pattern shared by intestinal E. coli. Infect Immun 63: 2070–2074, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.McLaughlin J, Padfield PJ, Burt JP, O’Neill CA. Ochratoxin A increases permeability through tight junctions by removal of specific claudin isoforms. Am J Physiol Cell Physiol 287: C1412–C1417, 2004. [DOI] [PubMed] [Google Scholar]
- 78.McNamara BP, Koutsouris A, O’Connell CB, Nougayrede JP, Donnenberg MS, Hecht G. Translocated EspF protein from enteropathogenic Escherichia coli disrupts host intestinal barrier function. J Clin Invest 107: 621–629, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Meza I, Sabanero M, Stefani E, Cereijido M. Occluding junctions in MDCK cells: Modulation of transepithelial permeability by the cytoskeleton. J Cell Biochem 18: 407–421, 1982. [DOI] [PubMed] [Google Scholar]
- 80.Mills E, Baruch K, Aviv G, Nitzan M, Rosenshine I. Dynamics of the type III secretion system activity of enteropathogenic Escherichia coli. MBio 4(4): pii: e00303–13, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Montecucco C, Rappuoli R. Living dangerously: How Helicobacter pylori survives in the human stomach. Nat Rev Mol Cell Biol 2: 457–466, 2001. [DOI] [PubMed] [Google Scholar]
- 82.Moore R, Pothoulakis C, LaMont JT, Carlson S, Madara JL. C. difficile toxin A increases intestinal permeability and induces Cl− secretion. Am J Physiol 259: G165–G172, 1990. [DOI] [PubMed] [Google Scholar]
- 83.Muza-Moons MM, Koutsouris A, Hecht G. Disruption of cell polarity by enteropathogenic Escherichia coli enables basolateral membrane proteins to migrate apically and to potentiate physiological consequences. Infect Immun 71: 7069–7078, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Muza-Moons MM, Schneeberger EE, Hecht GA. Enteropathogenic Escherichia coli infection leads to appearance of aberrant tight junctions strands in the lateral membrane of intestinal epithelial cells. Cell Microbiol 6: 783–793, 2004. [DOI] [PubMed] [Google Scholar]
- 85.Nava P, Kamekura R, Nusrat A. Cleavage of transmembrane junction proteins and their role in regulating epithelial homeostasis. Tissue Barriers 1: e24783, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Naydenov NG, Feygin A, Wang D, Kuemmerle JF, Harris G, Conti MA, Adelstein RS, Ivanov AI. Nonmuscle myosin IIA regulates intestinal epithelial barrier in vivo and plays a protective role during experimental colitis. Sci Rep 6: 24161, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Necchi V, Candusso ME, Tava F, Luinetti O, Ventura U, Fiocca R, Ricci V, Solcia E. Intracellular, intercellular, and stromal invasion of gastric mucosa, preneoplastic lesions, and cancer by Helicobacter pylori. Gastroenterology 132: 1009–1023, 2007. [DOI] [PubMed] [Google Scholar]
- 88.Nguyen M, Rizvi J, Hecht G. Expression of enteropathogenic Escherichia coli map is significantly different than that of other type III secreted effectors in vivo. Infect Immun 83: 130–137, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Noach LA, Rolf TM, Tytgat GN. Electron microscopic study of association between Helicobacter pylori and gastric and duodenal mucosa. J Clin Pathol 47: 699–704, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Nusrat A, Chen JA, Foley CS, Liang TW, Tom J, Cromwell M, Quan C, Mrsny RJ. The coiled-coil domain of occludin can act to organize structural and functional elements of the epithelial tight junction. J Biol Chem 275: 29816–29822, 2000. [DOI] [PubMed] [Google Scholar]
- 91.Papini E, Satin B, Norais N, de Bernard M, Telford JL, Rappuoli R, Montecucco C. Selective increase of the permeability of polarized epithelial cell monolayers by Helicobacter pylori vacuolating toxin. J Clin Invest 102: 813–820, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pelicic V, Reyrat JM, Sartori L, Pagliaccia C, Rappuoli R, Telford JL, Montecucco C, Papini E. Helicobacter pylori VacA cytotoxin associated with the bacteria increases epithelial permeability independently of its vacuolating activity. Microbiology 145 (Pt 8): 2043–2050, 1999. [DOI] [PubMed] [Google Scholar]
- 93.Peralta-Ramirez J, Hernandez JM, Manning-Cela R, Luna-Munoz J, Garcia-Tovar C, Nougayrede JP, Oswald E, Navarro-Garcia F. EspF Interacts with nucleation-promoting factors to recruit junctional proteins into pedestals for pedestal maturation and disruption of paracellular permeability. Infect Immun 76: 3854–3868, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Philpott DJ, McKay DM, Sherman PM, Perdue MH. Infection of T84 cells with enteropathogenic Escherichia coli alters barrier and transport functions. Am J Physiol 270: G634–G645, 1996. [DOI] [PubMed] [Google Scholar]
- 95.Polk DB, Peek RM Jr., Helicobacter pylori: Gastric cancer and beyond. Nat Rev Cancer 10: 403–414, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Popoff MR, Bouvet P. Clostridial toxins. Future Microbiol 4: 1021–1064, 2009. [DOI] [PubMed] [Google Scholar]
- 97.Quiros M, Nusrat A. RhoGTPases, actomyosin signaling and regulation of the epithelial apical junctional complex. Semin Cell Dev Biol 36: 194–203, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Raleigh DR, Boe DM, Yu D, Weber CR, Marchiando AM, Bradford EM, Wang Y, Wu L, Schneeberger EE, Shen L, Turner JR. Occludin S408 phosphorylation regulates tight junction protein interactions and barrier function. J Cell Biol 193: 565–582, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Rodriguez-Boulan E, Macara IG. Organization and execution of the epithelial polarity programme. Nat Rev Mol Cell Biol 15: 225–242, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Rossetto O, Pirazzini M, Montecucco C. Botulinum neurotoxins: Genetic, structural and mechanistic insights. Nat Rev Microbiol 12: 535–549, 2014. [DOI] [PubMed] [Google Scholar]
- 101.Rummel A The long journey of botulinum neurotoxins into the synapse. Toxicon 107: 9–24, 2015. [DOI] [PubMed] [Google Scholar]
- 102.Rupnik M, Wilcox MH, Gerding DN. Clostridium difficile infection: New developments in epidemiology and pathogenesis. Nat Rev Microbiol 7: 526–536, 2009. [DOI] [PubMed] [Google Scholar]
- 103.Sakaguchi G Clostridium botulinum toxins. Pharmacol Ther 19: 165–194, 1982. [DOI] [PubMed] [Google Scholar]
- 104.Sakaguchi T, Kohler H, Gu X, McCormick BA, Reinecker HC. Shigella flexneri regulates tight junction-associated proteins in human intestinal epithelial cells. Cell Microbiol 4: 367–381, 2002. [DOI] [PubMed] [Google Scholar]
- 105.Santos AS, Finlay BB. Bringing down the host: Enteropathogenic and enterohaemorrhagic Escherichia coli effector-mediated subversion of host innate immune pathways. Cell Microbiol 17: 318–332, 2015. [DOI] [PubMed] [Google Scholar]
- 106.Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junction pore and leak pathways: A dynamic duo. Annu Rev Physiol 73: 283–309, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Shifflett DE, Clayburgh DR, Koutsouris A, Turner JR, Hecht GA. Enteropathogenic E. coli disrupts tight junction barrier function and structure in vivo. Lab Invest 85: 1308–1324, 2005. [DOI] [PubMed] [Google Scholar]
- 108.Shin K, Fogg VC, Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol 22: 207–235, 2006. [DOI] [PubMed] [Google Scholar]
- 109.Shrestha A, Uzal FA, McClane BA. The interaction of Clostridium perfringens enterotoxin with receptor claudins. Anaerobe 41: 18–26, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Simonovic I, Arpin M, Koutsouris A, Falk-Krzesinski HJ, Hecht G. Enteropathogenic Escherichia coli activates ezrin, which participates in disruption of tight junction barrier function. Infect Immun 69: 5679–5688, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Simonovic I, Rosenberg J, Koutsouris A, Hecht G. Enteropathogenic Escherichia coli dephosphorylates and dissociates occludin from intestinal epithelial tight junctions. Cell Microbiol 2: 305–315, 2000. [DOI] [PubMed] [Google Scholar]
- 112.Singh AP, Aijaz S. Generation of a MDCK cell line with constitutive expression of the Enteropathogenic E. coli effector protein Map as an in vitro model of pathogenesis. Bioengineered 6: 335–341, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Singh U, Mitic LL, Wieckowski EU, Anderson JM, McClane BA. Comparative biochemical and immunocytochemical studies reveal differences in the effects of Clostridium perfringens enterotoxin on polarized CaCo-2 cells versus Vero cells. J Biol Chem 276: 33402–33412, 2001. [DOI] [PubMed] [Google Scholar]
- 114.Singh U, Van Itallie CM, Mitic LL, Anderson JM, McClane BA. CaCo-2 cells treated with Clostridium perfringens enterotoxin form multiple large complex species, one of which contains the tight junction protein occludin. J Biol Chem 275: 18407–18417, 2000. [DOI] [PubMed] [Google Scholar]
- 115.Smits WK, Lyras D, Lacy DB, Wilcox MH, Kuijper EJ. Clostridium difficile infection. Nat Rev Dis Primers 2: 16020, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Song X, Chen HX, Wang XY, Deng XY, Xi YX, He Q, Peng TL, Chen J, Chen W, Wong BC, Chen MH. H. pylori-encoded CagA disrupts tight junctions and induces invasiveness of AGS gastric carcinoma cells via Cdx2-dependent targeting of Claudin-2. Cell Immunol 286: 22–30, 2013. [DOI] [PubMed] [Google Scholar]
- 117.Sonoda N, Furuse M, Sasaki H, Yonemura S, Katahira J, Horiguchi Y, Tsukita S. Clostridium perfringens enterotoxin fragment removes specific claudins from tight junction strands: Evidence for direct involvement of claudins in tight junction barrier. J Cell Biol 147: 195–204, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Spindler V, Meir M, Vigh B, Flemming S, Hutz K, Germer CT, Waschke J, Schlegel N. Loss of desmoglein 2 contributes to the pathogenesis of Crohn’s disease. Inflamm Bowel Dis 21: 2349–2359, 2015. [DOI] [PubMed] [Google Scholar]
- 119.Spitz J, Yuhan R, Koutsouris A, Blatt C, Alverdy J, Hecht G. Enteropathogenic Escherichia coli adherence to intestinal epithelial monolayers diminishes barrier function. Am J Physiol 268: G374–G379, 1995. [DOI] [PubMed] [Google Scholar]
- 120.Stevenson BR, Anderson JM, Goodenough DA, Mooseker MS. Tight junction structure and ZO-1 content are identical in two strains of Madin-Darby canine kidney cells which differ in transepithelial resistance. J Cell Biol 107: 2401–2408, 1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sugawara Y, Matsumura T, Takegahara Y, Jin Y, Tsukasaki Y, Takeichi M, Fujinaga Y. Botulinum hemagglutinin disrupts the intercellular epithelial barrier by directly binding E-cadherin. J Cell Biol 189: 691–700, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Suzuki K, Kokai Y, Sawada N, Takakuwa R, Kuwahara K, Isogai E, Isogai H, Mori M. Helicobacter pylori disrupts the paracellular barrier of the gastric mucosa and leads to neutrophilic gastritis in mice. Virchows Arch 440: 318–324, 2002. [DOI] [PubMed] [Google Scholar]
- 123.Tafazoli F, Magnusson KE, Zheng L. Disruption of epithelial barrier integrity by Salmonella enterica serovar Typhimurium requires geranylgeranylated proteins. Infect Immun 71: 872–881, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Tai YH, Gage TP, McQueen C, Formal SB, Boedeker EC. Electrolyte transport in rabbit cecum. I. Effect of RDEC-1 infection. Am J Physiol 256: G721–G726, 1989. [DOI] [PubMed] [Google Scholar]
- 125.Tanaka-Okamoto M, Hori K, Ishizaki H, Itoh Y, Onishi S, Yonemura S, Takai Y, Miyoshi J. Involvement of afadin in barrier function and homeostasis of mouse intestinal epithelia. J Cell Sci 124: 2231–2240, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tanaka H, Yamamoto Y, Kashihara H, Yamazaki Y, Tani K, Fujiyoshi Y, Mineta K, Takeuchi K, Tamura A, Tsukita S. Claudin-21 has a paracellular channel role at tight junctions. Mol Cell Biol 36: 954–964, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tapia R, Kralicek SE, Hecht GA. EPEC effector EspF promotes Crumbs3 endocytosis and disrupts epithelial cell polarity. Cell Microbiol 19: e12757–n/a, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Tapia R, Kralicek SE, Hecht GA. Modulation of epithelial cell polarity by bacterial pathogens. Ann N Y Acad Sci 1405(1): 16–24, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Thanabalasuriar A, Koutsouris A, Hecht G, Gruenheid S. The bacterial virulence factor NleA’s involvement in intestinal tight junction disruption during enteropathogenic E. coli infection is independent of its putative PDZ binding domain. Gut Microbes 1: 114–118, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Tomson FL, Koutsouris A, Viswanathan VK, Turner JR, Savkovic SD, Hecht G. Differing roles of protein kinase C-zeta in disruption of tight junction barrier by enteropathogenic and enterohemorrhagic Escherichia coli. Gastroenterology 127: 859–869, 2004. [DOI] [PubMed] [Google Scholar]
- 131.Tomson FL, Viswanathan VK, Kanack KJ, Kanteti RP, Straub KV, Menet M, Kaper JB, Hecht G. Enteropathogenic Escherichia coli EspG disrupts microtubules and in conjunction with Orf3 enhances perturbation of the tight junction barrier. Mol Microbiol 56: 447–464, 2005. [DOI] [PubMed] [Google Scholar]
- 132.Tsai PY, Zhang B, He WQ, Zha JM, Odenwald MA, Singh G, Tamura A, Shen L, Sailer A, Yeruva S, Kuo WT, Fu YX, Tsukita S, Turner JR. IL-22 Upregulates epithelial claudin-2 to drive diarrhea and enteric pathogen clearance. Cell Host Microbe 21: 671–681 e674, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Turner HL, Turner JR. Good fences make good neighbors: Gastrointestinal mucosal structure. Gut Microbes 1: 22–29, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Turner JR, Buschmann MM, Romero-Calvo I, Sailer A, Shen L. The role of molecular remodeling in differential regulation of tight junction permeability. Semin Cell Dev Biol 36: 204–212, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Turner JR, Rill BK, Carlson SL, Carnes D, Kerner R, Mrsny RJ, Madara JL. Physiological regulation of epithelial tight junctions is associated with myosin light-chain phosphorylation. Am J Physiol 273: C1378–C1385, 1997. [DOI] [PubMed] [Google Scholar]
- 136.Ugalde-Silva P, Gonzalez-Lugo O, Navarro-Garcia F. Tight junction disruption induced by type 3 secretion system effectors injected by enteropathogenic and enterohemorrhagic Escherichia coli. Front Cell Infect Microbiol 6: 87, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ungewiss H, Vielmuth F, Suzuki ST, Maiser A, Harz H, Leonhardt H, Kugelmann D, Schlegel N, Waschke J. Desmoglein 2 regulates the intestinal epithelial barrier via p38 mitogen-activated protein kinase. Sci Rep 7: 6329, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Van Itallie CM, Holmes J, Bridges A, Gookin JL, Coccaro MR, Proctor W, Colegio OR, Anderson JM. The density of small tight junction pores varies among cell types and is increased by expression of claudin-2. J Cell Sci 121: 298–305, 2008. [DOI] [PubMed] [Google Scholar]
- 139.Viswanathan VK, Koutsouris A, Lukic S, Pilkinton M, Simonovic I, Simonovic M, Hecht G. Comparative analysis of EspF from enteropathogenic and enterohemorrhagic Escherichia coli in alteration of epithelial barrier function. Infect Immun 72: 3218–3227, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Viswanathan VK, Weflen A, Koutsouris A, Roxas JL, Hecht G. Enteropathogenic E. coli-induced barrier function alteration is not a consequence of host cell apoptosis. Am J Physiol Gastrointest Liver Physiol 294: G1165–G1170, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wang D, Naydenov NG, Feygin A, Baranwal S, Kuemmerle JF, Ivanov AI. Actin-depolymerizing factor and cofilin-1 have unique and overlapping functions in regulating intestinal epithelial junctions and mucosal inflammation. Am J Pathol 186: 844–858, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Watson CJ, Rowland M, Warhurst G. Functional modeling of tight junctions in intestinal cell monolayers using polyethylene glycol oligomers. Am J Physiol Cell Physiol 281: C388–C397, 2001. [DOI] [PubMed] [Google Scholar]
- 143.Watson PM, Paterson JC, Thom G, Ginman U, Lundquist S, Webster CI. Modelling the endothelial blood-CNS barriers: A method for the production of robust in vitro models of the rat blood-brain barrier and blood-spinal cord barrier. BMC Neurosci 14: 59, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Weber CR, Liang GH, Wang Y, Das S, Shen L, Yu AS, Nelson DJ, Turner JR. Claudin-2-dependent paracellular channels are dynamically gated. Elife 4: e09906, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Weber CR, Raleigh DR, Su L, Shen L, Sullivan EA, Wang Y, Turner JR. Epithelial myosin light chain kinase activation induces mucosal interleukin-13 expression to alter tight junction ion selectivity. J Biol Chem 285: 12037–12046, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Weflen AW, Alto NM, Hecht GA. Tight junctions and enteropathogenic E. coli. Ann N Y Acad Sci 1165: 169–174, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wong AR, Pearson JS, Bright MD, Munera D, Robinson KS, Lee SF, Frankel G, Hartland EL. Enteropathogenic and enterohaemorrhagic Escherichia coli: Even more subversive elements. Mol Microbiol 80: 1420–1438, 2011. [DOI] [PubMed] [Google Scholar]
- 148.Wroblewski LE, Piazuelo MB, Chaturvedi R, Schumacher M, Aihara E, Feng R, Noto JM, Delgado A, Israel DA, Zavros Y, Montrose MH, Shroyer N, Correa P, Wilson KT, Peek RM, Jr. Helicobacter pylori targets cancer-associated apical-junctional constituents in gastroids and gastric epithelial cells. Gut 64: 720–730, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Wroblewski LE, Shen L, Ogden S, Romero-Gallo J, Lapierre LA, Israel DA, Turner JR, Peek RM, Jr. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology 136: 236–246, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Yuhan R, Koutsouris A, Savkovic SD, Hecht G. Enteropathogenic Escherichia coli-induced myosin light chain phosphorylation alters intestinal epithelial permeability. Gastroenterology 113: 1873–1882, 1997. [DOI] [PubMed] [Google Scholar]
- 151.Zemljic M, Rupnik M, Scarpa M, Anderluh G, Palu G, Castagliuolo I. Repetitive domain of Clostridium difficile toxin B exhibits cytotoxic effects on human intestinal epithelial cells and decreases epithelial barrier function. Anaerobe 16: 527–532, 2010. [DOI] [PubMed] [Google Scholar]
- 152.Zhang Q, Li Q, Wang C, Li N, Li J. Redistribution of tight junction proteins during EPEC infection in vivo. Inflammation 35: 23–32, 2012. [DOI] [PubMed] [Google Scholar]
- 153.Zhang Y, Wu S, Ma J, Xia Y, Ai X, Sun J. Bacterial protein AvrA stabilizes intestinal epithelial tight junctions via blockage of the C-Jun N-terminal kinase pathway. Tissue Barriers 3: e972849, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zhang YG, Wu S, Xia Y, Sun J. Salmonella infection upregulates the leaky protein claudin-2 in intestinal epithelial cells. PLoS One 8: e58606, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zihni C, Mills C, Matter K, Balda MS. Tight junctions: From simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol 17: 564–580, 2016. [DOI] [PubMed] [Google Scholar]
- 156.Zolotarevsky Y, Hecht G, Koutsouris A, Gonzalez DE, Quan C, Tom J, Mrsny RJ, Turner JR. A membrane-permeant peptide that inhibits MLC kinase restores barrier function in in vitro models of intestinal disease. Gastroenterology 123: 163–172, 2002. [DOI] [PubMed] [Google Scholar]