Skip to main content
Cellular and Molecular Gastroenterology and Hepatology logoLink to Cellular and Molecular Gastroenterology and Hepatology
. 2020 Apr 15;10(2):327–340. doi: 10.1016/j.jcmgh.2020.04.001

Tight Junctions as Targets and Effectors of Mucosal Immune Homeostasis

Li Zuo 1,2,, Wei-Ting Kuo 2,, Jerrold R Turner 2,
PMCID: PMC7326733  PMID: 32304780

Abstract

Defective epithelial barrier function is present in maladies including epidermal burn injury, environmental lung damage, renal tubular disease, and a range of immune-mediated and infectious intestinal disorders. When the epithelial surface is intact, the paracellular pathway between cells is sealed by the tight junction. However, permeability of tight junctions varies widely across tissues and can be markedly impacted by disease. For example, tight junctions within the skin and urinary bladder are largely impermeant and their permeability is not regulated. In contrast, tight junctions of the proximal renal tubule and intestine are selectively permeable to water and solutes on the basis of their biophysical characteristics and, in the gut, can be regulated by the immune system with remarkable specificity. Conversely, modulation of tight junction barrier conductance, especially within the gastrointestinal tract, can impact immune homeostasis and diverse pathologies. Thus, tight junctions are both effectors and targets of immune regulation. Using the gastrointestinal tract as an example, this review explores current understanding of this complex interplay between tight junctions and immunity.

Keywords: Intestinal Permeability, Barrier, Myosin Light Chain Kinase, Enteric Infection, Inflammatory Bowel Disease, Graft-Versus-Host Disease, Pore Pathway, Leak Pathway, Claudin

Abbreviations used in this paper: B6, C57BL/6; BMT, bone marrow/hematopoietic stem cell transplantation; DSS, dextran sulfate sodium; GVHD, graft-versus-host disease; IBD, inflammatory bowel disease; IL, interleukin; JAM-A, junctional adhesion molecule-A; LIGHT, lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry mediator on T cells; MDCK, Madin-Darby canine kidney; MLC, myosin II regulatory light chain; MLCK, myosin light chain kinase; NK, natural killer; TER, transepithelial electrical resistance; TNF, tumor necrosis factor; ZO, zonula occludens


Summary.

Paracellular transport across the selectively permeable mucosal barrier is essential for health. Two distinct pathways, pore and leak, mediate transport across the tight junction, which is the rate-limiting step in paracellular flux. The permeabilities of these routes can be differentially regulated by immune and other stimuli and, conversely, have distinct effects on intestinal and systemic immune function.

Mucosal surfaces are lined by epithelial cells that, depending on the site, mediate and regulate nutrition absorption,1, 2, 3, 4, 5 secretion,5, 6, 7 physical barrier protection,8,9 transcellular transport,4,10 and environmental sensing.11, 12, 13, 14, 15 At these sites, the plasma membranes of epithelial cells, along with extracellular components (eg, mucin), establish a barrier that prevents free exchange of materials between the lumen and subepithelial tissues (ie, the lamina propria). Nevertheless, a potential shunt pathway between adjacent epithelial cells must also be sealed. This requires structural support by desmosomes and adherens junctions, which link epithelial cells to one another, and tight junctions, which limit paracellular flux. Importantly, tight junctions are not absolute seals, but are selectively permeable barriers that discriminate between water and solutes on the basis of size and charge. Two distinct pathways across the tight junction have been described and can be separately regulated by immune signals. Conversely, changes in the permeability of each pathway can differentially modulate mucosal immune activation. Thus, the interaction between tight junctions and mucosal immune system is a dynamic conversation with signals being transmitted in both directions. Some forms of immune activation and other stimuli reduce intestinal barrier function by directly damaging the epithelium, thereby creating a flux route termed the unrestricted pathway. Finally, any analysis of signaling between the immune system and epithelium must consider the means by which luminal materials, including microbiota and their metabolites, interact with the epithelium.

Impact of Mucosal Immune Regulation on Tight Junction Permeability

The complete molecular composition and structure of tight junctions remain to be defined. However, a great deal of progress has been made over the half-century since tight junctions were initially described.16, 17, 18 This includes discovery of zonula occludens (ZO)-119 and the related cytoplasmic scaffolding proteins ZO-220 and ZO-321; cingulin22; the tight junction associated Marvel proteins occludin,23 tricellulin,24 and marvelD325,26; claudins27, 28, 29, 30, 31; and others.32,33 Beyond these compositional proteins, the tight junction is functionally and structurally linked to the subcortical terminal web of actin microfilaments and the perijunctional actomyosin ring.34, 35, 36, 37, 38, 39

Solutes and water cross the tight junction by two distinct pathways that can be distinguished on the basis of their size-selectivity, charge-selectivity, and capacity (Figure 1A).40,41 The pore pathway is a high-conductance route that is charge-selective and extremely size-selective, with an upper limit of 6- to 8-Å diameter. In contrast, the less well-defined upper size limit of the lower conductance, charge nonselective leak pathway has been estimated to be ∼100-Å diameter.42 This model is consistent with in vivo studies of mucosal permeability along the villus-crypt axis, which identified distinct paracellular flux routes that could be distinguished on the basis of size-selectivity; that work concluded that 12-Å diameter pores were present in the villus but that larger, 100- to 120-Å diameter pores populated the crypts.43

Figure 1.

Figure 1

Mechanisms of paracellular permeability. (A) Two distinct routes are responsible for trans-tight junction flux. The pore pathway, whose permeability is primarily regulated by the specific claudin proteins expressed, is exquisitely size-selective and excludes molecules with diameters greater than 8 Å. The pore pathway is also charge selective. For example, claudin-2 specifically increases paracellular flux of cations (eg, Na+), and water. The leak pathway allows macromolecular flux and is thought to have an exclusion limit of ∼100 Å. In some inflammatory conditions, occludin down-regulation, including endocytic removal from the tight junction, leads to increased leak pathway permeability. A third route, the unrestricted pathway, describes flux at sites of epithelial damage and is tight junction-independent.109,127 (B) Ribbon diagram of claudins as viewed from the apical aspect of the tight junction. The pore formed by interactions between β-sheets within extracellular loop 1 of claudins on adjacent cells54,128,129 is indicated. (C) Space-filling model of the 3 α-helices formed by the coiled-coil occludin/ELL domain within the occludin cytoplasmic tail. Six adjacent lysines, including K433, form a basic (blue) ZO-1 binding interface (arrow).78,130,131

The Pore Pathway

The pore pathway was identified in parallel by 2 sets of experiments. Van Itallie et al44 analyzed flux of polyethylene glycols across pig ileum and monolayers of Caco-2 intestinal epithelial cells and 2 distinct clones of Madin-Darby canine kidney (MDCK) cells; all demonstrated a size-restrictive pore with a sharp size cutoff at ∼8-Å diameter. When the 2 MDCK lines, which had markedly different transepithelial electrical resistances (TERs), were compared, increased flux of 7-Å diameter polyethylene glycol correlated with increased ion conductance (ie, reduced TER). Analysis of tight junction protein expression showed that MDCK II, the MDCK line with greater polyethylene glycol flux, expressed claudin-2 but that the less permeable MDCK C7 line did not. Expression of claudin-2 in MDCK C7 cells reduced TER and enhanced paracellular flux of 7-Å diameter polyethylene glycol, but not larger polyethylene glycols, consistent with previous work showing that forced claudin-2 expression in MDCK C7 monolayers increased Na+, but not 4-kDa dextran, flux.30 Van Itallie et al44 therefore concluded that claudin-2 expression increased the number of small tight junction pores.

Concurrently, Weber et al45 treated T84 intestinal epithelial cell monolayers with interleukin (IL)-13 and found that this increased paracellular cation permeability but did not affect flux of 4-kDa dextran. Detailed study showed that IL-13 selectively induced claudin-2 expression and that siRNA-mediated blockade of claudin-2 up-regulation prevented IL-13-induced conductance increases.45 This confirmed observations in MDCK C7 cells, as described previously, and further demonstrated that IL-13 selectively enhances paracellular permeability by the high conductance, charge, and size-selective pore pathway.45

Further understanding of claudin-2-mediated pore pathway conductance was provided by a series of mutagenesis studies that identified specific residues that define the claudin-2 pore.46, 47, 48, 49 These were all within the first extracellular loop of claudin-2 (Figure 1B) and could be mapped to narrower and wider portions of the channel. Subsequent patch clamp analyses demonstrated that claudin-2 channels are actively gated and have single channel conductances of ∼9 pA.50 Together, these data indicate that, although claudin-2 channels are located between cells and are oriented parallel to plasma membranes, they have significant similarities to traditional transmembrane ion channels.

The data described above focus on claudin-2, a member of the claudin protein family. Alternative splicing of the 27 claudin genes allows expression of an even greater number of proteins. Individual claudin proteins are differentially expressed within specific tissues and cell types; the patterns of expression are also modified during development and in response to extracellular stimuli, including immune cells and their products. In general, claudin proteins have been subdivided into pore-forming and barrier-forming classes. Claudin-2 is a pore-forming claudin, as are claudins 10a, 10b, 15, 16, and 17; these form channels that are either cation- or anion-selective. Conversely, claudin- expression in MDCK II monolayers reduces paracellular flux of Na+ and 7-Å diameter polyethylene glycol. More detailed discussion of claudin proteins, their functions, and interactions are available.51, 52, 53, 54, 55, 56, 57, 58, 59

The tremendous efficacy of transmembrane ion channel inhibitors in many disorders suggests that development of specific means to modulate pore pathway tight junction channels may also be therapeutic. One approach to claudin-2 channel inhibition involves inhibition of casein kinase 2. This results in dephosphorylation of serine 408 within the C-terminal occludin tail and assembly of a tripartite complex composed of occludin, ZO-1, and claudin-2.60 Incorporation into this complex de-anchors claudin-2 at the tight junction and disrupts channel function. For example, casein kinase-2 inhibition acutely reversed IL-13-induced increases in paracellular permeability of T84 monolayers.60 Although translation to in vivo applications has not been reported and will likely require more specificity than casein kinase-2 inhibition provides,61 these data indicate that molecular targeting of protein interactions has the potential to modulate claudin channels and pore pathway permeability.

The Leak Pathway

In contrast to the pore pathway, the specific sites of leak pathway flux have not been defined. One possibility is that transient breaks within tight junction strands allow macromolecules (>8-Å diameter) to pass.62, 63, 64 This hypothesis proposes that, as strands reform, macromolecules are trapped in interstrand spaces until a break in the next strand allows them to continue to move across the tight junction. As discussed later, tricellular tight junctions, where 3 cells meet, have also been proposed as specialized sites of paracellular, macromolecular flux.65

Despite lack of structural understanding, components of the signal transduction machinery that regulates leak pathway permeability have been studied extensively.66 The most well-characterized of these is myosin light chain kinase (MLCK), which regulates paracellular permeability during physiological, Na+-nutrient cotransport.39,67 Expression of constitutively-active MLCK is sufficient to increase leak pathway permeability in vitro68 and in vivo.69

Based on the hypothesis that tight junction signaling mechanisms triggered by physiological stimuli mediate transduction by pathophysiological stimuli, Zolotarevsky et al70 asked if MLCK was involved in tight junction barrier loss induced by tumor necrosis factor (TNF). They showed that a highly specific MLCK inhibitor, PIK, was able to reverse both increased myosin II regulatory light chain (MLC) phosphorylation and reduced TER induced by TNF in vitro.70 Subsequent in vivo analyses demonstrated that increases in intestinal epithelial MLC phosphorylation paralleled fluid accumulation during acute T-cell activation-induced diarrhea.71 Pharmacologic or genetic intestinal epithelial MLCK inhibition prevented these TNF-induced increases in MLC phosphorylation, luminal fluid accumulation, and albumin (ie, leak pathway) permeability.71

Remarkably, the distribution of most tight junction proteins was unaffected by T-cell activation.71,72 Intestinal epithelial occludin was, however, internalized in a manner that correlated directly with intestinal barrier loss and could be blocked by MLCK inhibition (Figure 2). This TNF-induced occludin endocytosis occurred via caveolae and was prevented by caveolin-1 knockout, which blocked leak pathway permeability increases without affecting TNF-induced MLC phosphorylation (Figure 2). These data, therefore, established occludin endocytosis as a marker of TNF-induced, MLCK-mediated increases in leak pathway permeability. Nevertheless, the contributions of occludin to barrier function have been questioned.73,74 However, transgenic EGFP-occludin overexpression within the intestinal epithelium markedly attenuated TNF-induced increases in leak pathway permeability and restored net fluid absorption. Occludin is, therefore, a critical regulator of leak pathway permeability.72

Figure 2.

Figure 2

Acute, TNF-induced barrier loss is regulated by MLCK-dependent, caveolar occludin endocytosis. (A) Systemic T-cell activation, induced by anti-CD3 antibody treatment, causes acute, TNF-dependent diarrhea.71,80,132 After 3 hours, jejunal tissues were stained for occludin (green) and nuclei (blue). Anti-CD3 treatment induced occludin endocytosis in wild-type (WT) mice. In contrast, mice lacking long MLCK (MLCKKO) were resistant to anti-CD3-induced occludin endocytosis. (B) WT and Cav1-/- mice were injected with vehicle or recombinant TNF to induce diarrhea similar to that triggered by systemic T-cell activation. Images show jejunal tissues labeled for occludin (green), F-actin (red), and nuclei (blue). Caveolin-1 is required for TNF-induced occludin internalization. (C) Leak pathway permeability was assessed by blood-to-intestinal lumen flux of labeled albumin. Both anti-CD3 and TNF treatment increased leak pathway permeability. These increases were, however, blocked in MLCKKO and caveolin-1 knockout mice. (D) Anti-CD3- and TNF-induced leak pathway permeability increases correlated with reversal of net fluid flow from absorption to secretion. Water absorption was maintained in MLCKKO and caveolin-1 knockout mice. Data from Clayburgh et al71 and Marchiando et al.72

Although occludin knockout mice have been reported to have normal intestinal barrier function,73,74 male sterility and deafness suggest that occludin is critical to epithelial barrier function within the testes and cochlear hair cells.73,75 Consistent with this, occludin overexpression enhanced barrier function of MDCK monolayers,76 and studies of both MDCK and Caco-2 occludin knockdown lines demonstrated increased paracellular permeability to macromolecules with diameters up to ∼100 Å.42,77

Occludin-deficient MDCK and Caco-2 epithelial monolayers are resistant to TNF-induced barrier loss.42,72,76 This was not caused by a failure of signal transduction, because TNF-induced MLC phosphorylation was intact in occludin-deficient Caco-2 cells.42 Further analysis showed that the coiled-coil occludin/ELL domain within the cytoplasmic C-terminal occludin tail is required for TNF-induced permeability increases and that this depends on K433, which forms part of the occludin binding surface for ZO-1.42,78 This interaction with ZO-1 may be central to MLCK-dependent leak pathway regulation, because ZO-1, but not occludin, binds directly to F-actin.79 Consistent with this idea, the actin binding region of ZO-1 is required for in vitro barrier regulation by MLCK.37 Moreover, ZO-1 knockdown increases leak pathway permeability of epithelial monolayers.63

Similar to TNF, the TNF core family member LIGHT (lymphotoxin-like inducible protein that competes with glycoprotein D for herpes virus entry on T cells) and IL-1β trigger MLCK activation, occludin internalization, and increased leak pathway permeability in vitro and in vivo.80, 81, 82, 83, 84 In contrast to TNF, LIGHT did not cause net fluid secretion (ie, diarrhea). This difference reflects the ability of TNF, but not LIGHT, to down-regulate Na+ absorption by Na+/H+ exchanger isoform 3.80 Remarkably, ongoing Na+/H+ exchanger isoform 3–mediated transcellular Na+ transport supported increased fluid absorption in LIGHT-treated mice, demonstrating the passive nature of paracellular flux. In this case, the gradient created by Na+/H+ exchanger isoform 3–mediated Na+ absorption transport dictated the direction of paracellular water flow. Regulation intestinal and renal paracellular transport by transcellular transport and, conversely, support of transcellular transport by paracellular flux, have also been described in the absence of disease.85, 86, 87, 88

Beyond the focus on occludin and ZO-1, some authors have hypothesized that flux of macromolecules via the leak pathway occurs at tricellular junctions.65 This idea is consistent with the observation that tricellulin overexpression reduces paracellular macromolecular flux65 and morphologic analyses demonstrating a unique tight junction structure at tricellular contacts89,90 that is disrupted in tricellulin knockout mice.91 The observation that high-dose IL-13, 100- to 1000-fold greater than that required for claudin-2 up-regulation, reduced tricellulin expression, and increased 4-kDa dextran permeability could lend further support to the hypothesis that tricellulin seals the leak pathway.45,92 It must, however, be recognized that, at these doses, IL-13 can induce apoptosis.93

Contributions of tricellulin to increased leak pathway permeability may also be related to occludin endocytosis, because occludin loss causes tricellulin to expand its distribution to include bicellular tight junction regions.94,95 This relationship is made more complex by the observation that a tricellulin-derived peptide that displaces tricellulin from tight junctions and increases macromolecular paracellular flux also causes occludin internalization.96 The actin cytoskeleton, ZO-1, and at least 2 members of the tight junction associated Marvel proteins family, occludin and tricellulin, are therefore implicated in leak pathway regulation. Further work is needed to identify the anatomic sites and molecular mechanisms of leak pathway flux.

Impact of Tight Junction Permeability on Mucosal Immune Regulation

For many years conventional wisdom has dictated that increased intestinal permeability is a cause of disease. This belief was based on intuition; observations that massive barrier loss, such as the extensive epithelial damage caused by dextran sulfate sodium (DSS), could cause experimental colitis; and a correlation between disease severity and the magnitude of intestinal barrier loss in other disorders. However, data refuting this concept were reported more than 30 years ago. These data demonstrated that increased intestinal permeability was present in a subset of entirely healthy relatives of patients with Crohn’s disease.97 Despite being reproduced in many studies and linked specifically to NOD2 risk alleles,98,99 this documentation of increased intestinal permeability in healthy subjects has not received widespread recognition.

Studies in mice confirm the conclusion that intestinal barrier defects that fall short of substantial mucosal damage are insufficient to cause overt disease. These include analyses of junctional adhesion molecule-A (JAM-A; F11r) knockout mice demonstrating the absence of spontaneous disease despite increased intestinal permeability100,101 and increased intestinal epithelial proliferation (a sensitive marker of epithelial damage).102 Transgenic mice expressing constitutively-active MLCK within the intestinal epithelium were also healthy despite increased leak pathway permeability.69 Nevertheless, both JAM-A knockout and transgenic constitutively-active MLCK expression induced low-grade mucosal immune activation characterized by increased numbers of lamina propria CD4 T cells and IgA-producing plasma cells.69,100,103,104 JAM-A knockout mice were also hypersensitive to DSS-induced colitis,103 which was further exacerbated by elimination of transforming growth factor-β-producing CD4 T cells or knockout of the IgA heavy chain gene Igha.103 An adaptive immune response characterized by increased transforming growth factor-β and IgA production may therefore partially compensate for intestinal barrier loss as a consequence of JAM-A deletion. Constitutively-active MLCK-induced permeability increases also activated mucosal immunity that was sufficient to limit acute translocation of pathogenic bacteria (Salmonella typhimurium) and parasites (Toxoplasma gondii).104 This protection required a complex gut microbiome and IL-17-producing CD4 T cells but was not dependent on increased IgA production.104

Although these studies of mice with genetic defects demonstrate mucosal immune activation that partially compensates for intestinal barrier loss, recent detailed analyses of have identified more subtle changes. This includes alterations of the gut microbiome, behavior, visceral sensitivity, and neuronal activation within stress-response regions of the brain in constitutively-active MLCK transgenic mice.105 Although further study is needed, these data may be provisionally interpreted as evidence that modest increases in intestinal permeability can impact the gut-brain axis and trigger phenotypically diverse responses.

The MLCK-Regulated Leak Pathway as an Effector of Immune-Mediated Disease

In addition to the gut microbiome, T-cell transfer colitis depends on the absence of regulatory T cells.106,107 Thus, although outstanding for many purposes, the absence of regulatory T cells prevents this model from providing an unbiased picture of the evolution of mucosal immunity in chronic disease. To better define this, Nalle et al108, 109, 110 studied the contributions of intestinal barrier defects to development and progression of graft-versus-host disease (GVHD), a major complication of bone marrow/hematopoietic stem cell transplantation (BMT). At first, they focused on contributions of barrier defects to GVHD initiation.108

As in humans, BMT in mice requires preconditioning to eliminate the endogenous hematopoietic stem cells. In most mouse studies this is accomplished by irradiation, which damages the bone marrow and intestinal epithelium, two of the most rapidly proliferating cellular compartments. It is therefore not surprising that unrestricted pathway permeability was increased in the week after preconditioning. Intestinal permeability continued to increase after major antigen mismatch BMT from BALB/c donors into C57BL/6 (B6) recipients.108 In contrast, intestinal permeability normalized in mice receiving syngeneic or minor antigen mismatch BMT from B6 or 129S donors, respectively. After a lag period of several weeks, intestinal permeability then began to increase in mice that had received minor antigen mismatch BMT. Thus, major antigen mismatch GVHD was associated with a monophasic increase in intestinal permeability, whereas intestinal barrier defects were biphasic after minor antigen mismatch BMT.

Differentiation between increased intestinal permeability as a cause or effect of GVHD has not been possible, because the gut is a target of preconditioning damage and disease. To overcome this, Nalle et al108, 109, 110 used immunodeficient (Rag1-/-) mice as BMT recipients. Mice that received pre-BMT irradiation developed GVHD as expected, but neither minor antigen nor major antigen mismatch BMT was sufficient to cause GVHD in the absence of preconditioning. Flow cytometric analyses excluded rejection of donor T cells in nonirradiated mice as a trivial explanation for the lack of disease but did demonstrate that irradiation effectively cleared endogenous natural killer (NK) cells. Further study showed that recipient mice in which NK cell function had been eliminated by anti-NK antibody-mediated depletion or perforin knockout developed GVHD after major antigen mismatch BMT. Thus, intestinal barrier loss was not required for major antigen mismatch GVHD to develop.

In contrast to mice that received major antigen mismatch BMT, minor antigen mismatch BMT was unable to cause GVHD despite NK cell depletion or perforin knockout.108 This could be overcome by DSS pretreatment, to induce colonic damage, or by intraperitoneal lipopolysaccharide (ie, endotoxin) administration.108 Thus, intestinal damage, or at least systemic exposure to bacterial products, is required for the development of minor antigen mismatch GVHD.108 Moreover, low-grade GVHD developed in constitutively-active MLCK transgenic Rag1-/- mice that received minor antigen mismatch BMT and NK cell depletion without irradiation, DSS, or lipopolysaccharide (unpublished data, Nalle and Turner). Thus, initiation of GVHD following a modest immune stimulus (ie, minor antigen mismatch BMT) requires a second signal provided by intestinal barrier loss. This can be overcome by strong immune stimuli (ie, major antigen mismatch).108

The biphasic nature of barrier defects in minor antigen mismatch GVHD prompted further analysis.110 The first phase of intestinal permeability increases was caused by irradiation and mucosal damage. However, the second phase of barrier loss began in the interval between recovery from irradiation and development of clinically evident disease. By 2 weeks after BMT, intestinal epithelial MLC phosphorylation was markedly increased in mice that received minor antigen mismatch allogeneic BMT relative to those that received syngeneic BMT or control mice that were neither irradiated nor transplanted (Figure 3A).110 Increased MLC phosphorylation was associated with transcriptional MLCK up-regulation within intestinal epithelia, suggesting that that MLCK-dependent increases in tight junction permeability might be responsible for the second phase of barrier loss in mice. Consistent with this, intestinal permeability to 4 kDa dextran was increased at 5 weeks after allogeneic (minor antigen mismatch) BMT in B6 mice, but not in B6 mice lacking long (nonmuscle) MLCK.110 These MLCK knockout mice were also protected from GVHD overall on the basis of serum cytokine elevation (Figure 3B), histologic damage, weight loss (Figure 3C), and survival. Although long MLCK is expressed in other cell types, including endothelial cells, endothelial leakage persisted in the long MLCK knockout mice, indicating that vascular barriers were not protected by long MLCK knockout. More importantly, complementation of long MLCK knockout by intestinal epithelial-specific constitutively-active MLCK restored sensitivity to disease, thereby demonstrating that intestinal epithelial MLCK is critical to disease progression. The observation that MLCK expression and MLC phosphorylation are increased in intestinal epithelia of patients with GVHD, relative to healthy control subjects, suggests that the same mechanisms of leak pathway regulation contribute to pathogenesis of human disease.110

Figure 3.

Figure 3

Intestinal epithelial MLCK-dependent leak pathway permeability increases drive systemic GVHD. (A) Phosphorylated MLC (green) within intestinal epithelial cells (red) is markedly increased following allogeneic (129→B6), but not syngeneic (B6→B6) BMT. Intestinal epithelial MLC phosphorylation is not increased following allogeneic BMT into long MLCK knockout recipients. Smooth muscle MLC phosphorylation within the villus core is unaffected by long MLCK knockout. (B) Increases in serum TNF levels following allogeneic BMT are attenuated in long MLCK knockout BMT recipients. (C) Long MLCK knockout mice are protected from GVHD-associated weight loss. (D) Intestinal CD8 T cell (green) infiltration is markedly reduced in long MLCK knockout BMT recipients. (E) Model showing that intestinal barrier loss during GVHD can be separated into three distinct phases: (1) conditioning-induced epithelial damage and unrestricted pathway permeability increases (initiation), (2) cytokine-induced tight junction leak pathway regulation (propagation), and (3) T cell–mediated tissue damage and increased unrestricted pathway permeability (advanced disease). Data from Nalle et al.110 ECAD, E-cadherin; pMLC, phosphorylated MLC; WT, wild-type.

Tissue analysis showed that, in addition to reduced damage, infiltration by terminally differentiated cytolytic (CD8+/granzyme B+) T cells was markedly reduced in MLCK knockout allogeneic BMT recipients (Figure 3D). To determine whether these were antigen-specific, pathogenic T cells or mere bystanders, a different GVHD model, using B6 transgenic mice expressing membrane-bound ovalbumin on the surface of all cells was used. These mice received a syngeneic BMT that included a small number of splenocytes from OT-I transgenic mice, whose CD8 T cells recognize ovalbumin.110 When analyzed in mesenteric lymph nodes, granzyme B expression within antigen-specific (OT-I) CD8 T cells was markedly reduced in MLCK knockout, relative to wild-type, BMT recipients. In contrast, granzyme B expression in OT-I CD8 T cells from nonmesenteric peripheral lymph nodes and spleen was similar in wild-type and long MLCK knockout mice. This indicates that MLCK-dependent intestinal barrier loss promotes local, terminal differentiation of antigen-specific T cells during evolution of GVHD. Remarkably, analysis of the nonantigen-specific CD8 T cells also showed reduced numbers with granzyme B expression in mesenteric lymph nodes, but not other sites, in long MLCK knockout mice. Therefore, intestinal barrier loss drives GVHD progression by promoting terminal differentiation of polyclonal populations that include antigen-specific and nonantigen-specific, cytolytic CD8 T cells (Figure 3E).

These data suggest that MLCK-mediated, leak pathway barrier loss may make similar contributions to immune activation in other diseases, such as inflammatory bowel disease (IBD), in which pathogenesis is not driven by a single antigen. Consistent with this, immune-mediated experimental IBD (T-cell transfer colitis) was more severe in constitutively-active MLCK transgenic mice.69 Conversely, knockout mice lacking long MLCK were protected from experimental IBD.111 As in experimental GVHD, this protection was eliminated by intestinal epithelial-specific expression of constitutively-active MLCK.111 However, in contrast to GVHD, experimental IBD ultimately progressed in the MLCK knockout mice. Onset of disease in the knockout mice correlated temporally with intestinal permeability loss caused by epithelial apoptosis.111 These data indicate that intestinal barrier loss is critical to evolution of experimental IBD and that, like GVHD, disease amelioration by inhibition of MLCK-mediated leak pathway permeability increases may be overcome by strong immune stimuli.

Although these data suggest that MLCK inhibition might be an effective therapy in immune-mediated intestinal disease, it is important to remember that MLCK serves other critical epithelial functions, such as migration and wound repair.112,113 Moreover, because the gene that encodes epithelial MLCK also encodes smooth muscle MLCK, any enzymatic inhibitor of epithelial MLCK would inhibit smooth muscle contraction and cause hypotension and intestinal obstruction.114 Finally, available MLCK enzymatic inhibitors are unable to discriminate between nonmuscle, smooth muscle, skeletal muscle, and cardiac MLCK isoforms. Thus, enzymatic MLCK inhibition is not a feasible approach to therapy. However, recent work has shown that a specific epithelial MLCK splice variant, long MLCK1, contains an unique domain that is required for effective recruitment to the perijunctional actomyosin ring and MLCK-dependent leak pathway regulation.38 A small molecule inhibitor that blocks long MLCK1 recruitment to the perijunctional actomyosin ring and prevents subsequent increases in leak pathway permeability without inhibiting MLCK enzymatic activity has recently been described.38 This molecule, termed Divertin, because it diverts long MLCK1 from the perijunctional actomyosin ring, was remarkably effective in a variety of in vitro and in vivo IBD models and, in T cell transfer colitis, was more effective than anti-TNF.38 Although only a single report of molecule that has not undergone complete pharmacologic analysis, this striking result suggests that it may be possible to target the leak pathway without systemic toxicity.

Consequences of Pore Pathway Regulation in Disease

Several studies have linked claudin-2 expression to MLCK-dependent barrier regulation.45,111,115 For example, constitutively-active MLCK expression within the intestinal epithelium led to increased claudin-2 expression and cation-selective, pore pathway permeability increases.45 This may have been caused by increased IL-13 production in constitutively-active MLCK transgenic mice. Furthermore, in experimental IBD, claudin-2 up-regulation was blocked in MLCK knockout mice but restored by complementation with constitutively-active MLCK.111 Finally, a study of claudin-2 knockout mice reported increased TNF-induced nuclear factor-κB activation and MLCK transcription in vivo.115 One interpretation of these data could be that claudin-2 is a downstream effector of MLCK-induced barrier loss in disease. Unfortunately, studies of immune-mediated colitis in claudin-2-deficient mice have not been reported.

Intestinal epithelial claudin-2 expression can be increased by IL-13, IL-22, IL-6, and a broad range of other stimuli.92,115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125 The impact of claudin-2 up-regulation on disease progression is, however, incompletely defined. One study showed that transgenic mice overexpressing human claudin-2 within the intestinal epithelium were protected from DSS-induced colitis.126 These mice, however, had other abnormalities, including marked up-regulation of epithelial proliferation, consistent with damage, and increased permeability to 4-kDa dextran, which cannot be represent flux across claudin-2 channels.126 Moreover, the mechanism of this protection may relate more to increased water content and reduced DSS concentration in the distal colon than a specific effect of claudin-2. Consistent with this, fecal water and Na+ were increased in a different transgenic mouse expressing EGFP-tagged mouse claudin-2.118 Conversely, DSS colitis is more severe in claudin-2 knockout mice.115

Claudin-2 knockout and intestinal epithelial-specific transgenic mice have been studied carefully in the context of infectious colitis.118 This investigation was prompted by results of in vivo, size-specific permeability assays, using creatinine (6-Å diameter), 4-kDa dextran (28-Å diameter), and 70-kDa dextran (120-Å diameter), that showed increased pore pathway permeability within 2 days of Citrobacter rodentium infection (Figure 4A and B). Leak pathway and unrestricted pathway permeabilities were increased at later times. Among all claudins, only claudin-2 expression was up-regulated within 2 days of infection (Figure 4C). This was associated with increased mucosal IL-22 but no changes in other cytokines. In vitro analysis of organoid cultures demonstrated that IL-22 was able to specifically up-regulate claudin-2. Moreover, IL-22 neutralizing antibodies prevented claudin-2 up-regulation at this early time after infection.118

Figure 4.

Figure 4

IL-22-induced claudin-2 up-regulation increases pore pathway permeability to promote intestinal pathogen clearance. (A) Flux of creatinine (6-Å diameter), 4-kDa dextran (28-Å diameter), and 70-kDa dextran (120-Å diameter) were used to assess pore, leak, and unrestricted pathway permeabilities, respectively. Intestinal permeability to these probes increased sequentially during the course of infection. (B) After normalization to 70-kDa dextran flux, a marker of unrestricted pathway permeability, pore and leak pathway probes demonstrated specific up-regulation at 2 and 3 days after infection, respectively. (C) Initial increases in intestinal epithelial claudin-2 (green) expression correlated with increased pore pathway permeability. F-actin (red) and DNA (blue) are shown for reference. (D) A model showing that Citrobacter rodentium infection elicits IL-22 release that leads to claudin-2 up-regulation, water and Na+ efflux, and pathogen clearance. Data abstracted from Tsai et al.118

To better understand the impact of claudin-2 up-regulation on infectious colitis, wild-type, claudin-2 knockout, and claudin-2 transgenic mice were compared. C rodentium–induced colitis was far more severe in claudin-2 knockout mice, as demonstrated by analyses of weight loss, tissue damage, proinflammatory cytokine expression, and numbers of mucosal-adherent bacteria. Fecal C rodentium shedding was prolonged in claudin-2 knockout mice, suggesting that claudin-2 promotes pathogen clearance.118 To test the hypothesis that claudin-2 primarily drives pathogen clearance by facilitating paracellular water and Na+ efflux into the lumen, polyethylene glycol was added to the drinking water of all 3 genotypes. Because polyethylene glycol cannot be absorbed, this creates an osmotic force that draws water and Na+ into the colonic lumen. This maneuver rescued claudin-2 knockout mice such that their disease was similar in magnitude to that of wild-type or claudin-2 transgenic mice, as assessed by histopathology, cytokine production, and numbers of mucosa-associated C rodentium.118 The protection afforded by claudin-2 up-regulation therefore depends on claudin-2-mediated water efflux (Figure 4D). How this water efflux promotes pathogen clearance has yet to be determined. It also remains to be determined whether increased claudin-2 expression impacts progression of inflammatory disorders, such as IBD.

Conclusions

There has been a tremendous expansion of the understanding of tight junction permeability, the biophysics of distinct tight junction flux pathways, and regulatory mechanisms responsible for tight junction regulation in recent years. The field is also beginning to realize the long sought-after goal of therapeutically modulating tight junction barrier function. Although many challenges remain, the next few years promise extraordinary advances.

Acknowledgments

The authors thank Ms. Tiffany S. Davanzo, CMI, for her outstanding illustrations. They also thank previous laboratory members who have graciously allowed their data to be reformatted and presented here, acknowledge the outstanding contributions of others in this field, and apologize to those whose work they were unable to cite because of space limitations. Li Zuo: conceptualization, equal; funding acquisition, supporting; writing original draft, lead; writing review and editing, equal. Wei-Ting Kuo: conceptualization, equal; writing original draft, equal; writing review and editing, equal. Jerrold R. Turner: conceptualization, lead; funding acquisition, lead; writing original draft, lead.

Footnotes

Conflicts of interest This author discloses the following: Jerrold R. Turner is a co-founder of Thelium Therapeutics, Inc. The remaining authors disclose no conflicts.

Funding This work was supported by NIH grants R01DK61931 (JRT), R01DK68271 (JRT), and R24DK099803 (JRT); the Harvard Digestive Disease Center (P30DK034854); the Department of Defense CDMRP PR181271 (JRT); and by National Natural Science Foundation of China grant 81800464 (LZ).

References

  • 1.Chen L., Tuo B., Dong H. Regulation of intestinal glucose absorption by ion channels and transporters. Nutrients. 2016;8 doi: 10.3390/nu8010043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kato A., Romero M.F. Regulation of electroneutral NaCl absorption by the small intestine. Annu Rev Physiol. 2011;73:261–281. doi: 10.1146/annurev-physiol-012110-142244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kellett G.L., Brot-Laroche E., Mace O.J., Leturque A. Sugar absorption in the intestine: the role of GLUT2. Annu Rev Nutr. 2008;28:35–54. doi: 10.1146/annurev.nutr.28.061807.155518. [DOI] [PubMed] [Google Scholar]
  • 4.Pappenheimer J.R. On the coupling of membrane digestion with intestinal absorption of sugars and amino acids. Am J Physiol. 1993;265:G409–G417. doi: 10.1152/ajpgi.1993.265.3.G409. [DOI] [PubMed] [Google Scholar]
  • 5.Turnberg L.A. Absorption and secretion of salt and water by the small intestine. Digestion. 1973;9:357–381. doi: 10.1159/000197461. [DOI] [PubMed] [Google Scholar]
  • 6.Binder H.J., Rajendran V., Sadasivan V., Geibel J.P. Bicarbonate secretion: a neglected aspect of colonic ion transport. J Clin Gastroenterol. 2005;39:S53–S58. doi: 10.1097/01.mcg.0000155521.81382.3a. [DOI] [PubMed] [Google Scholar]
  • 7.Barrett K.E., Keely S.J. Chloride secretion by the intestinal epithelium: molecular basis and regulatory aspects. Annu Rev Physiol. 2000;62:535–572. doi: 10.1146/annurev.physiol.62.1.535. [DOI] [PubMed] [Google Scholar]
  • 8.Liang G.H., Weber C.R. Molecular aspects of tight junction barrier function. Curr Opin Pharmacol. 2014;19C:84–89. doi: 10.1016/j.coph.2014.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Koch S., Nusrat A. Dynamic regulation of epithelial cell fate and barrier function by intercellular junctions. Ann N Y Acad Sci. 2009;1165:220–227. doi: 10.1111/j.1749-6632.2009.04025.x. [DOI] [PubMed] [Google Scholar]
  • 10.Khanal R.C., Nemere I. Regulation of intestinal calcium transport. Annu Rev Nutr. 2008;28:179–196. doi: 10.1146/annurev.nutr.010308.161202. [DOI] [PubMed] [Google Scholar]
  • 11.Smith K., Karimian Azari E., LaMoia T.E., Hussain T., Vargova V., Karolyi K., Veldhuis P.P., Arnoletti J.P., de la Fuente S.G., Pratley R.E., Osborne T.F., Kyriazis G.A. T1R2 receptor-mediated glucose sensing in the upper intestine potentiates glucose absorption through activation of local regulatory pathways. Mol Metab. 2018;17:98–111. doi: 10.1016/j.molmet.2018.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wang S., Charbonnier L.M., Noval Rivas M., Georgiev P., Li N., Gerber G., Bry L., Chatila T.A. MyD88 adaptor-dependent microbial sensing by regulatory T cells promotes mucosal tolerance and enforces commensalism. Immunity. 2015;43:289–303. doi: 10.1016/j.immuni.2015.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Knoop K.A., McDonald K.G., McCrate S., McDole J.R., Newberry R.D. Microbial sensing by goblet cells controls immune surveillance of luminal antigens in the colon. Mucosal Immunol. 2015;8:198–210. doi: 10.1038/mi.2014.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Oh J.Z., Ravindran R., Chassaing B., Carvalho F.A., Maddur M.S., Bower M., Hakimpour P., Gill K.P., Nakaya H.I., Yarovinsky F., Sartor R.B., Gewirtz A.T., Pulendran B. TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity. 2014;41:478–492. doi: 10.1016/j.immuni.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pacheco A.R., Curtis M.M., Ritchie J.M., Munera D., Waldor M.K., Moreira C.G., Sperandio V. Fucose sensing regulates bacterial intestinal colonization. Nature. 2012;492:113–117. doi: 10.1038/nature11623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Farquhar M., Palade G. Junctional complexes in various epithelia. J Cell Biol. 1963;17:375–412. doi: 10.1083/jcb.17.2.375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Staehelin L.A., Mukherjee T.M., Williams A.W. Freeze-etch appearance of the tight junctions in the epithelium of small and large intestine of mice. Protoplasma. 1969;67:165–184. doi: 10.1007/BF01248737. [DOI] [PubMed] [Google Scholar]
  • 18.Machen T.E., Erlij D., Wooding F.B. Permeable junctional complexes. The movement of lanthanum across rabbit gallbladder and intestine. J Cell Biol. 1972;54:302–312. doi: 10.1083/jcb.54.2.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Stevenson B.R., Siliciano J.D., Mooseker M.S., Goodenough D.A. Identification of ZO-1: a high molecular weight polypeptide associated with the tight junction (zonula occludens) in a variety of epithelia. J Cell Biol. 1986;103:755–766. doi: 10.1083/jcb.103.3.755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jesaitis L.A., Goodenough D.A. Molecular characterization and tissue distribution of ZO-2, a tight junction protein homologous to ZO-1 and the Drosophila discs-large tumor suppressor protein. J Cell Biol. 1994;124:949–961. doi: 10.1083/jcb.124.6.949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Haskins J., Gu L., Wittchen E.S., Hibbard J., Stevenson B.R. ZO-3, a novel member of the MAGUK protein family found at the tight junction, interacts with ZO-1 and occludin. J Cell Biol. 1998;141:199–208. doi: 10.1083/jcb.141.1.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Citi S., Sabanay H., Jakes R., Geiger B., Kendrick-Jones J. Cingulin, a new peripheral component of tight junctions. Nature. 1988;333:272–276. doi: 10.1038/333272a0. [DOI] [PubMed] [Google Scholar]
  • 23.Furuse M., Hirase T., Itoh M., Nagafuchi A., Yonemura S., Tsukita S., Tsukita S. Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol. 1993;123:1777–1788. doi: 10.1083/jcb.123.6.1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ikenouchi J., Furuse M., Furuse K., Sasaki H., Tsukita S., Tsukita S. Tricellulin constitutes a novel barrier at tricellular contacts of epithelial cells. J Cell Biol. 2005;171:939–945. doi: 10.1083/jcb.200510043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Steed E., Rodrigues N.T., Balda M.S., Matter K. Identification of MarvelD3 as a tight junction-associated transmembrane protein of the occludin family. BMC Cell Biol. 2009;10:95. doi: 10.1186/1471-2121-10-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Raleigh D.R., Marchiando A.M., Zhang Y., Shen L., Sasaki H., Wang Y., Long M., Turner J.R. Tight junction-associated MARVEL proteins marveld3, tricellulin, and occludin have distinct but overlapping functions. Mol Biol Cell. 2010;21:1200–1213. doi: 10.1091/mbc.E09-08-0734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Furuse M., Fujita K., Hiiragi T., Fujimoto K., Tsukita S. Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol. 1998;141:1539–1550. doi: 10.1083/jcb.141.7.1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.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. 2001;153:263–272. doi: 10.1083/jcb.153.2.263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Van Itallie C., Rahner C., Anderson J.M. Regulated expression of claudin-4 decreases paracellular conductance through a selective decrease in sodium permeability. J Clin Invest. 2001;107:1319–1327. doi: 10.1172/JCI12464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Amasheh S., Meiri N., Gitter A.H., Schoneberg T., Mankertz J., Schulzke J.D., Fromm M. Claudin-2 expression induces cation-selective channels in tight junctions of epithelial cells. J Cell Sci. 2002;115:4969–4976. doi: 10.1242/jcs.00165. [DOI] [PubMed] [Google Scholar]
  • 31.Mineta K., Yamamoto Y., Yamazaki Y., Tanaka H., Tada Y., Saito K., Tamura A., Igarashi M., Endo T., Takeuchi K., Tsukita S. Predicted expansion of the claudin multigene family. FEBS Lett. 2011;585:606–612. doi: 10.1016/j.febslet.2011.01.028. [DOI] [PubMed] [Google Scholar]
  • 32.Higashi T., Tokuda S., Kitajiri S., Masuda S., Nakamura H., Oda Y., Furuse M. Analysis of the ‘angulin’ proteins LSR, ILDR1 and ILDR2--tricellulin recruitment, epithelial barrier function and implication in deafness pathogenesis. J Cell Sci. 2013;126:966–977. doi: 10.1242/jcs.116442. [DOI] [PubMed] [Google Scholar]
  • 33.Kolosov D., Kelly S.P. Tricellular tight junction-associated angulins in the gill epithelium of rainbow trout. Am J Physiol Regul Integr Comp Physiol. 2018;315:R312–R322. doi: 10.1152/ajpregu.00431.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bentzel C.J., Hainau B., Edelman A., Anagnostopoulos T., Benedetti E.L. Effect of plant cytokinins on microfilaments and tight junction permeability. Nature. 1976;264:666–668. doi: 10.1038/264666a0. [DOI] [PubMed] [Google Scholar]
  • 35.Hull B.E., Staehelin L.A. The terminal web. A reevaluation of its structure and function. J Cell Biol. 1979;81:67–82. doi: 10.1083/jcb.81.1.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Madara J.L. Intestinal absorptive cell tight junctions are linked to cytoskeleton. Am J Physiol. 1987;253:C171–C175. doi: 10.1152/ajpcell.1987.253.1.C171. [DOI] [PubMed] [Google Scholar]
  • 37.Yu D., Marchiando A.M., Weber C.R., Raleigh D.R., Wang Y., Shen L., Turner J.R. MLCK-dependent exchange and actin binding region-dependent anchoring of ZO-1 regulate tight junction barrier function. Proc Natl Acad Sci U S A. 2010;107:8237–8241. doi: 10.1073/pnas.0908869107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Graham W.V., He W., Marchiando A.M., Zha J., Singh G., Li H.S., Biswas A., Ong M., Jiang Z.H., Choi W., Zuccola H., Wang Y., Griffith J., Wu J., Rosenberg H.J., Wang Y., Snapper S.B., Ostrov D., Meredith S.C., Miller L.W., Turner J.R. Intracellular MLCK1 diversion reverses barrier loss to restore mucosal homeostasis. Nat Med. 2019;25:690–700. doi: 10.1038/s41591-019-0393-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Turner J.R., Rill B.K., Carlson S.L., Carnes D., Kerner R., Mrsny R.J., Madara J.L. Physiological regulation of epithelial tight junctions is associated with myosin light-chain phosphorylation. Am J Physiol. 1997;273:C1378–C1385. doi: 10.1152/ajpcell.1997.273.4.C1378. [DOI] [PubMed] [Google Scholar]
  • 40.Turner J.R. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9:799–809. doi: 10.1038/nri2653. [DOI] [PubMed] [Google Scholar]
  • 41.Anderson J.M., Van Itallie C.M. Physiology and function of the tight junction. Cold Spring Harb Perspect Biol. 2009;1:a002584. doi: 10.1101/cshperspect.a002584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Buschmann M.M., Shen L., Rajapakse H., Raleigh D.R., Wang Y., Wang Y., Lingaraju A., Zha J., Abbott E., McAuley E.M., Breskin L.A., Wu L., Anderson K., Turner J.R., Weber C.R. Occludin OCEL-domain interactions are required for maintenance and regulation of the tight junction barrier to macromolecular flux. Mol Biol Cell. 2013;24:3056–3068. doi: 10.1091/mbc.E12-09-0688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fihn B.M., Sjöqvist A., Jodal M. Permeability of the rat small intestinal epithelium along the villus-crypt axis: effects of glucose transport. Gastroenterology. 2000;119:1029–1036. doi: 10.1053/gast.2000.18148. [DOI] [PubMed] [Google Scholar]
  • 44.Van Itallie C.M., Holmes J., Bridges A., Gookin J.L., Coccaro M.R., Proctor W., Colegio O.R., Anderson J.M. The density of small tight junction pores varies among cell types and is increased by expression of claudin-2. J Cell Sci. 2008;121:298–305. doi: 10.1242/jcs.021485. [DOI] [PubMed] [Google Scholar]
  • 45.Weber C.R., Raleigh D.R., Su L., Shen L., Sullivan E.A., Wang Y., Turner J.R. Epithelial myosin light chain kinase activation induces mucosal interleukin-13 expression to alter tight junction ion selectivity. J Biol Chem. 2010;285:12037–12046. doi: 10.1074/jbc.M109.064808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Angelow S., Yu A.S. Cysteine mutagenesis to study the structure of claudin-2 paracellular pores. Ann N Y Acad Sci. 2009;1165:143–147. doi: 10.1111/j.1749-6632.2009.04038.x. [DOI] [PubMed] [Google Scholar]
  • 47.Li J., Angelow S., Linge A., Zhuo M., Yu A.S. Claudin-2 pore function requires an intramolecular disulfide bond between two conserved extracellular cysteines. Am J Physiol Cell Physiol. 2013;305:C190–C196. doi: 10.1152/ajpcell.00074.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Li J., Zhuo M., Pei L., Yu A.S. Conserved aromatic residue confers cation selectivity in claudin-2 and claudin-10b. J Biol Chem. 2013;288:22790–22797. doi: 10.1074/jbc.M113.484238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yu A.S., Cheng M.H., Angelow S., Gunzel D., Kanzawa S.A., Schneeberger E.E., Fromm M., Coalson R.D. Molecular basis for cation selectivity in claudin-2-based paracellular pores: identification of an electrostatic interaction site. J Gen Physiol. 2009;133:111–127. doi: 10.1085/jgp.200810154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Weber C.R., Liang G.H., Wang Y., Das S., Shen L., Yu A.S., Nelson D.J., Turner J.R. Claudin-2-dependent paracellular channels are dynamically gated. Elife. 2015;4 doi: 10.7554/eLife.09906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gunzel D., Fromm M. Claudins and other tight junction proteins. Compr Physiol. 2012;2:1819–1852. doi: 10.1002/cphy.c110045. [DOI] [PubMed] [Google Scholar]
  • 52.Gunzel D., Yu A.S. Claudins and the modulation of tight junction permeability. Physiol Rev. 2013;93:525–569. doi: 10.1152/physrev.00019.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Tsukita S., Tanaka H., Tamura A. The claudins: from tight junctions to biological systems. Trends Biochem Sci. 2019;44:141–152. doi: 10.1016/j.tibs.2018.09.008. [DOI] [PubMed] [Google Scholar]
  • 54.Suzuki H., Tani K., Fujiyoshi Y. Crystal structures of claudins: insights into their intermolecular interactions. Ann N Y Acad Sci. 2017;1397:25–34. doi: 10.1111/nyas.13371. [DOI] [PubMed] [Google Scholar]
  • 55.Rosenthal R., Gunzel D., Theune D., Czichos C., Schulzke J.D., Fromm M. Water channels and barriers formed by claudins. Ann N Y Acad Sci. 2017;1397:100–109. doi: 10.1111/nyas.13383. [DOI] [PubMed] [Google Scholar]
  • 56.Garcia-Hernandez V., Quiros M., Nusrat A. Intestinal epithelial claudins: expression and regulation in homeostasis and inflammation. Ann N Y Acad Sci. 2017;1397:66–79. doi: 10.1111/nyas.13360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.France M.M., Turner J.R. The mucosal barrier at a glance. J Cell Sci. 2017;130:307–314. doi: 10.1242/jcs.193482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Liu F., Koval M., Ranganathan S., Fanayan S., Hancock W.S., Lundberg E.K., Beavis R.C., Lane L., Duek P., McQuade L., Kelleher N.L., Baker M.S. Systems proteomics view of the endogenous human claudin protein family. J Proteome Res. 2016;15:339–359. doi: 10.1021/acs.jproteome.5b00769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gunzel D. Claudins: vital partners in transcellular and paracellular transport coupling. Pflugers Arch. 2017;469:35–44. doi: 10.1007/s00424-016-1909-3. [DOI] [PubMed] [Google Scholar]
  • 60.Raleigh D.R., Boe D.M., Yu D., Weber C.R., Marchiando A.M., Bradford E.M., Wang Y., Wu L., Schneeberger E.E., Shen L., Turner J.R. Occludin S408 phosphorylation regulates tight junction protein interactions and barrier function. J Cell Biol. 2011;193:565–582. doi: 10.1083/jcb.201010065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Koch S., Capaldo C.T., Hilgarth R.S., Fournier B., Parkos C.A., Nusrat A. Protein kinase CK2 is a critical regulator of epithelial homeostasis in chronic intestinal inflammation. Mucosal Immunol. 2013;6:136–145. doi: 10.1038/mi.2012.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sasaki H., Matsui C., Furuse K., Mimori-Kiyosue Y., Furuse M., Tsukita S. Dynamic behavior of paired claudin strands within apposing plasma membranes. Proc Natl Acad Sci U S A. 2003;100:3971–3976. doi: 10.1073/pnas.0630649100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Van Itallie C.M., Fanning A.S., Bridges A., Anderson J.M. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol Biol Cell. 2009;20:3930–3940. doi: 10.1091/mbc.E09-04-0320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Van Itallie C.M., Tietgens A.J., Anderson J.M. Visualizing the dynamic coupling of claudin strands to the actin cytoskeleton through ZO-1. Mol Biol Cell. 2017;28:524–534. doi: 10.1091/mbc.E16-10-0698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Krug S.M., Amasheh S., Richter J.F., Milatz S., Gunzel D., Westphal J.K., Huber O., Schulzke J.D., Fromm M. Tricellulin forms a barrier to macromolecules in tricellular tight junctions without affecting ion permeability. Mol Biol Cell. 2009;20:3713–3724. doi: 10.1091/mbc.E09-01-0080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Buckley A., Turner J.R. Cell biology of tight junction barrier regulation and mucosal disease. Cold Spring Harb Perspect Biol. 2018;10 doi: 10.1101/cshperspect.a029314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Atisook K., Carlson S., Madara J.L. Effects of phlorizin and sodium on glucose-elicited alterations of cell junctions in intestinal epithelia. Am J Physiol. 1990;258:C77–C85. doi: 10.1152/ajpcell.1990.258.1.C77. [DOI] [PubMed] [Google Scholar]
  • 68.Shen L., Black E.D., Witkowski E.D., Lencer W.I., Guerriero V., Schneeberger E.E., Turner J.R. Myosin light chain phosphorylation regulates barrier function by remodeling tight junction structure. J Cell Sci. 2006;119:2095–2106. doi: 10.1242/jcs.02915. [DOI] [PubMed] [Google Scholar]
  • 69.Su L., Shen L., Clayburgh D.R., Nalle S.C., Sullivan E.A., Meddings J.B., Abraham C., Turner J.R. Targeted epithelial tight junction dysfunction causes immune activation and contributes to development of experimental colitis. Gastroenterology. 2009;136:551–563. doi: 10.1053/j.gastro.2008.10.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zolotarevsky Y., Hecht G., Koutsouris A., Gonzalez D.E., Quan C., Tom J., Mrsny R.J., Turner J.R. A membrane-permeant peptide that inhibits MLC kinase restores barrier function in in vitro models of intestinal disease. Gastroenterology. 2002;123:163–172. doi: 10.1053/gast.2002.34235. [DOI] [PubMed] [Google Scholar]
  • 71.Clayburgh D.R., Barrett T.A., Tang Y., Meddings J.B., Van Eldik L.J., Watterson D.M., Clarke L.L., Mrsny R.J., Turner J.R. Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo. J Clin Invest. 2005;115:2702–2715. doi: 10.1172/JCI24970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Marchiando A.M., Shen L., Graham W.V., Weber C.R., Schwarz B.T., Austin J.R., 2nd, Raleigh D.R., Guan Y., Watson A.J., Montrose M.H., Turner J.R. Caveolin-1-dependent occludin endocytosis is required for TNF-induced tight junction regulation in vivo. J Cell Biol. 2010;189:111–126. doi: 10.1083/jcb.200902153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Saitou M., Furuse M., Sasaki H., Schulzke J.D., Fromm M., Takano H., Noda T., Tsukita S. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol Biol Cell. 2000;11:4131–4142. doi: 10.1091/mbc.11.12.4131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schulzke J.D., Gitter A.H., Mankertz J., Spiegel S., Seidler U., Amasheh S., Saitou M., Tsukita S., Fromm M. Epithelial transport and barrier function in occludin-deficient mice. Biochim Biophys Acta. 2005;1669:34–42. doi: 10.1016/j.bbamem.2005.01.008. [DOI] [PubMed] [Google Scholar]
  • 75.Kitajiri S., Katsuno T., Sasaki H., Ito J., Furuse M., Tsukita S. Deafness in occludin-deficient mice with dislocation of tricellulin and progressive apoptosis of the hair cells. Biol Open. 2014;3:759–766. doi: 10.1242/bio.20147799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Van Itallie C.M., Fanning A.S., Holmes J., Anderson J.M. Occludin is required for cytokine-induced regulation of tight junction barriers. J Cell Sci. 2010;123:2844–2852. doi: 10.1242/jcs.065581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yu A.S., McCarthy K.M., Francis S.A., McCormack J.M., Lai J., Rogers R.A., Lynch R.D., Schneeberger E.E. Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells. Am J Physiol Cell Physiol. 2005;288:C1231–C1241. doi: 10.1152/ajpcell.00581.2004. [DOI] [PubMed] [Google Scholar]
  • 78.Tash B.R., Bewley M.C., Russo M., Keil J.M., Griffin K.A., Sundstrom J.M., Antonetti D.A., Tian F., Flanagan J.M. The occludin and ZO-1 complex, defined by small angle X-ray scattering and NMR, has implications for modulating tight junction permeability. Proc Natl Acad Sci U S A. 2012;109:10855–10860. doi: 10.1073/pnas.1121390109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Fanning A.S., Ma T.Y., Anderson J.M. Isolation and functional characterization of the actin binding region in the tight junction protein ZO-1. FASEB J. 2002;16:1835–1837. doi: 10.1096/fj.02-0121fje. [DOI] [PubMed] [Google Scholar]
  • 80.Clayburgh D.R., Musch M.W., Leitges M., Fu Y.X., Turner J.R. Coordinated epithelial NHE3 inhibition and barrier dysfunction are required for TNF-mediated diarrhea in vivo. J Clin Invest. 2006;116:2682–2694. doi: 10.1172/JCI29218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Schwarz B.T., Wang F., Shen L., Clayburgh D.R., Su L., Wang Y., Fu Y.X., Turner J.R. LIGHT signals directly to intestinal epithelia to cause barrier dysfunction via cytoskeletal and endocytic mechanisms. Gastroenterology. 2007;132:2383–2394. doi: 10.1053/j.gastro.2007.02.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Al-Sadi R.M., Ma T.Y. IL-1beta causes an increase in intestinal epithelial tight junction permeability. J Immunol. 2007;178:4641–4649. doi: 10.4049/jimmunol.178.7.4641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Al-Sadi R., Ye D., Dokladny K., Ma T.Y. Mechanism of IL-1beta-induced increase in intestinal epithelial tight junction permeability. J Immunol. 2008;180:5653–5661. doi: 10.4049/jimmunol.180.8.5653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Al-Sadi R., Guo S., Ye D., Dokladny K., Alhmoud T., Ereifej L., Said H.M., Ma T.Y. Mechanism of IL-1beta modulation of intestinal epithelial barrier involves p38 kinase and activating transcription factor-2 activation. J Immunol. 2013;190:6596–6606. doi: 10.4049/jimmunol.1201876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pei L., Solis G., Nguyen M.T., Kamat N., Magenheimer L., Zhuo M., Li J., Curry J., McDonough A.A., Fields T.A., Welch W.J., Yu A.S. Paracellular epithelial sodium transport maximizes energy efficiency in the kidney. J Clin Invest. 2016;126:2509–2518. doi: 10.1172/JCI83942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Meddings J.B., Westergaard H. Intestinal glucose transport using perfused rat jejunum in vivo: model analysis and derivation of corrected kinetic constants. Clin Sci (Lond) 1989;76:403–413. doi: 10.1042/cs0760403. [DOI] [PubMed] [Google Scholar]
  • 87.Simon D.B., Lu Y., Choate K.A., Velazquez H., Al-Sabban E., Praga M., Casari G., Bettinelli A., Colussi G., Rodriguez-Soriano J., McCredie D., Milford D., Sanjad S., Lifton R.P. Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science. 1999;285:103–106. doi: 10.1126/science.285.5424.103. [DOI] [PubMed] [Google Scholar]
  • 88.Wada M., Tamura A., Takahashi N., Tsukita S. Loss of claudins 2 and 15 from mice causes defects in paracellular Na+ flow and nutrient transport in gut and leads to death from malnutrition. Gastroenterology. 2013;144:369–380. doi: 10.1053/j.gastro.2012.10.035. [DOI] [PubMed] [Google Scholar]
  • 89.Staehelin L.A. Further observations on the fine structure of freeze-cleaved tight junctions. J Cell Sci. 1973;13:763–786. doi: 10.1242/jcs.13.3.763. [DOI] [PubMed] [Google Scholar]
  • 90.Walker D.C., MacKenzie A., Hulbert W.C., Hogg J.C. A re-assessment of the tricellular region of epithelial cell tight junctions in trachea of guinea pig. Acta Anat (Basel) 1985;122:35–38. doi: 10.1159/000145982. [DOI] [PubMed] [Google Scholar]
  • 91.Nayak G., Lee S.I., Yousaf R., Edelmann S.E., Trincot C., Van Itallie C.M., Sinha G.P., Rafeeq M., Jones S.M., Belyantseva I.A., Anderson J.M., Forge A., Frolenkov G.I., Riazuddin S. Tricellulin deficiency affects tight junction architecture and cochlear hair cells. J Clin Invest. 2013;123:4036–4049. doi: 10.1172/JCI69031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Krug S.M., Bojarski C., Fromm A., Lee I.M., Dames P., Richter J.F., Turner J.R., Fromm M., Schulzke J.D. Tricellulin is regulated via interleukin-13-receptor alpha2, affects macromolecule uptake, and is decreased in ulcerative colitis. Mucosal Immunol. 2018;11:345–356. doi: 10.1038/mi.2017.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Heller F., Fromm A., Gitter A.H., Mankertz J., Schulzke J.D. Epithelial apoptosis is a prominent feature of the epithelial barrier disturbance in intestinal inflammation: effect of pro-inflammatory interleukin-13 on epithelial cell function. Mucosal Immunol. 2008;1(Suppl 1):S58–S61. doi: 10.1038/mi.2008.46. [DOI] [PubMed] [Google Scholar]
  • 94.Ikenouchi J., Sasaki H., Tsukita S., Furuse M., Tsukita S. Loss of occludin affects tricellular localization of tricellulin. Mol Biol Cell. 2008;19:4687–4693. doi: 10.1091/mbc.E08-05-0530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Westphal J.K., Dorfel M.J., Krug S.M., Cording J.D., Piontek J., Blasig I.E., Tauber R., Fromm M., Huber O. Tricellulin forms homomeric and heteromeric tight junctional complexes. Cell Mol Life Sci. 2010;67:2057–2068. doi: 10.1007/s00018-010-0313-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Cording J., Arslan B., Staat C., Dithmer S., Krug S.M., Kruger A., Berndt P., Gunther R., Winkler L., Blasig I.E., Haseloff R.F. Trictide, a tricellulin-derived peptide to overcome cellular barriers. Ann N Y Acad Sci. 2017;1405:89–101. doi: 10.1111/nyas.13392. [DOI] [PubMed] [Google Scholar]
  • 97.Hollander D., Vadheim C.M., Brettholz E., Petersen G.M., Delahunty T., Rotter J.I. Increased intestinal permeability in patients with Crohn's disease and their relatives. A possible etiologic factor. Ann Intern Med. 1986;105:883–885. doi: 10.7326/0003-4819-105-6-883. [DOI] [PubMed] [Google Scholar]
  • 98.May G.R., Sutherland L.R., Meddings J.B. Is small intestinal permeability really increased in relatives of patients with Crohn's disease? Gastroenterology. 1993;104:1627–1632. doi: 10.1016/0016-5085(93)90638-s. [DOI] [PubMed] [Google Scholar]
  • 99.Buhner S., Buning C., Genschel J., Kling K., Herrmann D., Dignass A., Kuechler I., Krueger S., Schmidt H.H., Lochs H. Genetic basis for increased intestinal permeability in families with Crohn's disease: role of CARD15 3020insC mutation? Gut. 2006;55:342–347. doi: 10.1136/gut.2005.065557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Laukoetter M.G., Nava P., Lee W.Y., Severson E.A., Capaldo C.T., Babbin B.A., Williams I.R., Koval M., Peatman E., Campbell J.A., Dermody T.S., Nusrat A., Parkos C.A. JAM-A regulates permeability and inflammation in the intestine in vivo. J Exp Med. 2007;204:3067–3076. doi: 10.1084/jem.20071416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Vetrano S., Rescigno M., Cera M.R., Correale C., Rumio C., Doni A., Fantini M., Sturm A., Borroni E., Repici A., Locati M., Malesci A., Dejana E., Danese S. Unique role of junctional adhesion molecule-A in maintaining mucosal homeostasis in inflammatory bowel disease. Gastroenterology. 2008;135:173–184. doi: 10.1053/j.gastro.2008.04.002. [DOI] [PubMed] [Google Scholar]
  • 102.Nava P., Capaldo C.T., Koch S., Kolegraff K., Rankin C.R., Farkas A.E., Feasel M.E., Li L., Addis C., Parkos C.A., Nusrat A. JAM-A regulates epithelial proliferation through Akt/beta-catenin signalling. EMBO Rep. 2011;12:314–320. doi: 10.1038/embor.2011.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Khounlotham M., Kim W., Peatman E., Nava P., Medina-Contreras O., Addis C., Koch S., Fournier B., Nusrat A., Denning T.L., Parkos C.A. Compromised intestinal epithelial barrier induces adaptive immune compensation that protects from colitis. Immunity. 2012;37:563–573. doi: 10.1016/j.immuni.2012.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Edelblum K.L., Sharon G., Singh G., Odenwald M.A., Sailer A., Cao S., Ravens S., Thomsen I., El Bissati K., McLeod R., Dong C., Gurbuxani S., Prinz I., Mazmanian S.K., Turner J.R. The microbiome activates CD4 T-cell-mediated immunity to compensate for increased intestinal permeability. Cell Mol Gastroenterol Hepatol. 2017;4:285–297. doi: 10.1016/j.jcmgh.2017.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Inczefi O., Bacquie V., Olier-Pierre M., Rincel M., Ringot-Destrez B., Ellero-Simatos S., Eutamene H., Betoulieres C., Thomas J., Laine J., Gros L., Leveque M., Leonard R., Harkat C., Robbe-Masselot C., Roka R., Mercier-Bonin M., Theodorou V., Darnaudery M., Turner J.R., Ferrier L. Targeted intestinal tight junction hyperpermeability alters the microbiome, behavior, and visceromotor responses. Cell Mol Gastroenterol Hepatol. 2020 doi: 10.1016/j.jcmgh.2020.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Powrie F., Correa-Oliveira R., Mauze S., Coffman R.L. Regulatory interactions between CD45RBhigh and CD45RBlow CD4+ T cells are important for the balance between protective and pathogenic cell-mediated immunity. J Exp Med. 1994;179:589–600. doi: 10.1084/jem.179.2.589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Asseman C., Read S., Powrie F. Colitogenic Th1 cells are present in the antigen-experienced T cell pool in normal mice: control by CD4+ regulatory T cells and IL-10. J Immunol. 2003;171:971–978. doi: 10.4049/jimmunol.171.2.971. [DOI] [PubMed] [Google Scholar]
  • 108.Nalle S.C., Kwak H.A., Edelblum K.L., Joseph N.E., Singh G., Khramtsova G.F., Mortenson E.D., Savage P.A., Turner J.R. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Sci Transl Med. 2014;6:243ra87. doi: 10.1126/scitranslmed.3008941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Nalle S.C., Turner J.R. Intestinal barrier loss as a critical pathogenic link between inflammatory bowel disease and graft-versus-host disease. Mucosal Immunol. 2015;8:720–730. doi: 10.1038/mi.2015.40. [DOI] [PubMed] [Google Scholar]
  • 110.Nalle S.C., Zuo L., Ong M., Singh G., Worthylake A.M., Choi W., Manresa M.C., Southworth A.P., Edelblum K.L., Baker G.J., Joseph N.E., Savage P.A., Turner J.R. Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability. J Clin Invest. 2019;129:902–914. doi: 10.1172/JCI98554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Su L., Nalle S.C., Shen L., Turner E.S., Singh G., Breskin L.A., Khramtsova E.A., Khramtsova G., Tsai P.Y., Fu Y.X., Abraham C., Turner J.R. TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis. Gastroenterology. 2013;145:407–415. doi: 10.1053/j.gastro.2013.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Russo J.M., Florian P., Shen L., Graham W.V., Tretiakova M.S., Gitter A.H., Mrsny R.J., Turner J.R. Distinct temporal-spatial roles for rho kinase and myosin light chain kinase in epithelial purse-string wound closure. Gastroenterology. 2005;128:987–1001. doi: 10.1053/j.gastro.2005.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tamada M., Perez T.D., Nelson W.J., Sheetz M.P. Two distinct modes of myosin assembly and dynamics during epithelial wound closure. J Cell Biol. 2007;176:27–33. doi: 10.1083/jcb.200609116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.He W.Q., Peng Y.J., Zhang W.C., Lv N., Tang J., Chen C., Zhang C.H., Gao S., Chen H.Q., Zhi G., Feil R., Kamm K.E., Stull J.T., Gao X., Zhu M.S. Myosin light chain kinase is central to smooth muscle contraction and required for gastrointestinal motility in mice. Gastroenterology. 2008;135:610–620. doi: 10.1053/j.gastro.2008.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Nishida M., Yoshida M., Nishiumi S., Furuse M., Azuma T. Claudin-2 regulates colorectal inflammation via myosin light chain kinase-dependent signaling. Dig Dis Sci. 2013;58:1546–1559. doi: 10.1007/s10620-012-2535-3. [DOI] [PubMed] [Google Scholar]
  • 116.Heller F., Florian P., Bojarski C., Richter J., Christ M., Hillenbrand B., Mankertz J., Gitter A.H., Burgel N., Fromm M., Zeitz M., Fuss I., Strober W., Schulzke J.D. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology. 2005;129:550–564. doi: 10.1016/j.gastro.2005.05.002. [DOI] [PubMed] [Google Scholar]
  • 117.Tian L., Li Y., Zhang J., Chang R., Li J., Huo L. IL-9 promotes the pathogenesis of ulcerative colitis through STAT3/SOCS3 signaling. Biosci Rep. 2018;38 doi: 10.1042/BSR20181521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tsai P.Y., Zhang B., He W.Q., Zha J.M., Odenwald M.A., Singh G., Tamura A., Shen L., Sailer A., Yeruva S., Kuo W.T., Fu Y.X., Tsukita S., Turner J.R. IL-22 upregulates epithelial claudin-2 to drive diarrhea and enteric pathogen clearance. Cell Host Microbe. 2017;21:671–681. doi: 10.1016/j.chom.2017.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Wang Y., Mumm J.B., Herbst R., Kolbeck R., Wang Y. IL-22 increases permeability of intestinal epithelial tight junctions by enhancing claudin-2 expression. J Immunol. 2017;199:3316–3325. doi: 10.4049/jimmunol.1700152. [DOI] [PubMed] [Google Scholar]
  • 120.Suzuki T., Yoshinaga N., Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286:31263–31271. doi: 10.1074/jbc.M111.238147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Capaldo C.T., Farkas A.E., Hilgarth R.S., Krug S.M., Wolf M.F., Benedik J.K., Fromm M., Koval M., Parkos C., Nusrat A. Proinflammatory cytokine-induced tight junction remodeling through dynamic self-assembly of claudins. Mol Biol Cell. 2014;25:2710–2719. doi: 10.1091/mbc.E14-02-0773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ishimoto H., Oshima T., Sei H., Yamasaki T., Kondo T., Tozawa K., Tomita T., Ohda Y., Fukui H., Watari J., Miwa H. Claudin-2 expression is upregulated in the ileum of diarrhea predominant irritable bowel syndrome patients. J Clin Biochem Nutr. 2017;60:146–150. doi: 10.3164/jcbn.16-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Weber C.R., Nalle S.C., Tretiakova M., Rubin D.T., Turner J.R. Claudin-1 and claudin-2 expression is elevated in inflammatory bowel disease and may contribute to early neoplastic transformation. Lab Invest. 2008;88:1110–1120. doi: 10.1038/labinvest.2008.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wu R.L., Vazquez-Roque M.I., Carlson P., Burton D., Grover M., Camilleri M., Turner J.R. Gluten-induced symptoms in diarrhea-predominant irritable bowel syndrome are associated with increased myosin light chain kinase activity and claudin-15 expression. Lab Invest. 2017;97:14–23. doi: 10.1038/labinvest.2016.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhang Y.G., Wu S., Lu R., Zhou D., Zhou J., Carmeliet G., Petrof E., Claud E.C., Sun J. Tight junction CLDN2 gene is a direct target of the vitamin D receptor. Sci Rep. 2015;5:10642. doi: 10.1038/srep10642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ahmad R., Chaturvedi R., Olivares-Villagomez D., Habib T., Asim M., Shivesh P., Polk D.B., Wilson K.T., Washington M.K., Van Kaer L., Dhawan P., Singh A.B. Targeted colonic claudin-2 expression renders resistance to epithelial injury, induces immune suppression, and protects from colitis. Mucosal Immunol. 2014;7:1340–1353. doi: 10.1038/mi.2014.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Shen L., Weber C.R., Raleigh D.R., Yu D., Turner J.R. Tight junction pore and leak pathways: a dynamic duo. Annu Rev Physiol. 2011;73:283–309. doi: 10.1146/annurev-physiol-012110-142150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Suzuki H., Nishizawa T., Tani K., Yamazaki Y., Tamura A., Ishitani R., Dohmae N., Tsukita S., Nureki O., Fujiyoshi Y. Crystal structure of a claudin provides insight into the architecture of tight junctions. Science. 2014;344:304–307. doi: 10.1126/science.1248571. [DOI] [PubMed] [Google Scholar]
  • 129.Suzuki H., Tani K., Tamura A., Tsukita S., Fujiyoshi Y. Model for the architecture of claudin-based paracellular ion channels through tight junctions. J Mol Biol. 2015;427:291–297. doi: 10.1016/j.jmb.2014.10.020. [DOI] [PubMed] [Google Scholar]
  • 130.Li Y., Fanning A.S., Anderson J.M., Lavie A. Structure of the conserved cytoplasmic C-terminal domain of occludin: identification of the ZO-1 binding surface. J Mol Biol. 2005;352:151–164. doi: 10.1016/j.jmb.2005.07.017. [DOI] [PubMed] [Google Scholar]
  • 131.Lye M.F., Fanning A.S., Su Y., Anderson J.M., Lavie A. Insights into regulated ligand binding sites from the structure of ZO-1 Src homology 3-guanylate kinase module. J Biol Chem. 2010;285:13907–13917. doi: 10.1074/jbc.M109.093674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Musch M.W., Clarke L.L., Mamah D., Gawenis L.R., Zhang Z., Ellsworth W., Shalowitz D., Mittal N., Efthimiou P., Alnadjim Z., Hurst S.D., Chang E.B., Barrett T.A. T cell activation causes diarrhea by increasing intestinal permeability and inhibiting epithelial Na+/K+-ATPase. J Clin Invest. 2002;110:1739–1747. doi: 10.1172/JCI15695. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Gastroenterology and Hepatology are provided here courtesy of Elsevier

RESOURCES