Skip to main content

Intestinal barrier dysfunction in severe burn injury

Abstract

Severe burn injury is often accompanied by intestinal barrier dysfunction, which is closely associated with post-burn shock, bacterial translocation, systemic inflammatory response syndrome, hypercatabolism, sepsis, multiple organ dysfunction syndrome, and other complications. The intestinal epithelium forms a physical barrier that separates the intestinal lumen from the internal milieu, in which the tight junction plays a principal role. It has been well documented that after severe burn injury, many factors such as stress, ischemia/hypoxia, proinflammatory cytokines, and endotoxins can induce intestinal barrier dysfunction via multiple signaling pathways. Recent advances have provided new insights into the mechanisms and the therapeutic strategies of intestinal epithelial barrier dysfunction associated with severe burn injury. In this review, we will describe the current knowledge of the mechanisms involved in intestinal barrier dysfunction in response to severe burn injury and the emerging therapies for treating intestinal barrier dysfunction following severe burn injury.

Background

It is well known that the polarized epithelial cells of intestinal mucosa form a barrier to prevent luminal pathogens and antigenic molecules from entering the intestinal mucosa and contacting with the immune system, to which the tight junction (TJ) and its associated proteins are critical. However, the intestinal epithelial barrier disruption often develops in many diseases including severe burn injury. The severe burn-induced disruption of intestinal barrier results in the increased intestinal permeability and subsequent translocation of bacteria and/or endotoxin from the gastrointestinal tract to cause systemic inflammatory response syndrome, sepsis, multiple organ dysfunction syndrome, and other critical complications. Previous studies have shown a close relationship between intestinal barrier disruption and the incidence and severity of sepsis in critically ill patients [1, 2]. It has been demonstrated that 60% of critically ill patients suffering from multiple organ failure develop intestinal barrier disruption [2]. The burn victims occurring intestinal barrier disruption are at high risk for bacterial translocation, sepsis, and mortality [3]. Thus, the intestinal barrier disruption may play a central role in the development of multiple critical complications elicited by severe burn injury. Although the underlying mechanisms of intestinal barrier disruption induced by severe burn injury are not well understood, intensive research efforts regarding post-burn intestinal barrier dysfunction have been ongoing. The overall goal of this review is to describe the intestinal barrier disruption in severe burn injury, with a focus on the potential molecular mechanisms.

Review

Overview of intestinal barrier and TJ

The basic and important function of the intestinal tract is to digest and absorb nutrients. In addition, the intestinal tract is not only the largest immune organ in the body, but also the largest reservoir of bacteria and endotoxin. Under physiological condition, the intestinal mucosa allows only small molecules to pass, which relies on the intact mucosa barrier to effectively prevent luminal bacteria, endotoxins, and other antigens from translocating to other distant organs [4,5,6]. The intestinal barrier includes mechanical, immunological, biological, and chemical barriers. The mechanical barrier is mainly composed of the mucous layer on the surface of the intestinal mucosa, intestinal epithelial cells, intercellular junctions, submucosal lamina propria, etc. If there is no specific transporter, most hydrophilic solutes cannot permeate the mechanical barrier [5, 6]. The immunological barrier, which mainly consists of a large number of immunocompetent cells, including lymphocytes, macrophages, dendritic cells, and plasma cells, has the functions of antigen presentation, inflammatory mediators secretion, and mucosal allergic response [7]. The secretory IgA-mediated humoral immune response plays an important role in the intestinal immunological barrier. Lactobacillus and Bifidobacterium, which are the intestinal resident bacteria, mainly form the biological barrier. The resident bacteria form biofilm on the intestinal epithelial surfaces, resist the invasion of exogenous pathogenic bacteria, and provide the intestinal epithelial cells with nutrients by producing short-chain fatty acids, lactic acids, and others. The chemical barrier is the generic name for the antibacterial substances produced by the resident intestinal bacteria and chemical substances such as gastric acid, bile, digestive enzymes, muramidase, and mucopolysaccharide, which can inhibit the adherence and colonization of bacteria [8].

The mechanical barrier is a pivotal part of the intestinal barrier and maintained through the intestinal epithelial cells and intercellular junctions [4,5,6]. The intercellular junctions comprise the tight, gap, adhesion, and desmosome junctions. The TJ, consisting of multiple proteins such as zonula occludens (ZO), occludin, the claudins, and the junctional adhesion molecules (JAM), is a complex that forms a selectively permeable seal between adjacent epithelial cells and serves as a selective barrier between the plasma membranes of adjacent cells [9,10,11]. The TJ is in a state of relatively stable dynamic remodeling and regulated by various factors such as Ca2+-E-cadherin, Rho-GTPase, phospholipase C, protein kinase A, tyrosine kinase, mitogen-activated protein kinases (MAPK), and myosin light chain kinase (MLCK). Among these factors, MLCK plays a critical role in the regulation of TJ dynamic. By regulating the phosphorylation of myosin light chain (MLC), MLCK is associated with the perijunctional actomyosin ring [5].

Occludin, belonging to transmembrane proteins, is composed of four transmembrane domains, two extracellular domains, and a long cytoplasmic C-terminal tail [12]. The intracellular C-terminus of occludin is connected with ZO. Occludin has important significance in maintaining the paracellular permeability and transepithelial electricity [13,14,15]. The function of occludin depends on the phosphorylation of serine and threonine residues. The small GTPase- and protein kinase c (PKC)-related signaling pathways are involved in the regulation of occludin phosphorylation.

The claudin family contains more than 20 members, which have an extracellular loop structure of variable amino acid residues and an intracellular short tail structure [16,17,18]. Different subtypes of claudins are expressed in different tissues and cells. Claudins are not only the barrier-forming components of the TJ. There are also pore-forming claudins including claudin-2 and claudin-10. Thus, claudins play a critical role in the physical regulation of paracellular permeability by forming the intercellular ion channel [16].

ZO is a member of the so-called membrane-associated guanylate kinase (MAGUK) family and has the isomers ZO-1, ZO-2, and ZO-3 [19,20,21,22,23]. ZOs contain conserved sequences such as guanylate kinase (GUK) structure domains, PDZ domains, the Src-homologous SH3 domain, the acidic domain, the arginine-rich domain and the proline-rich domain, and these conserved sequences take part in the connection between ZO and other proteins. For example, ZO directly binds to the C-terminus of claudins through possynaptic density protein-95/Discs-Large/zonula occludens-1 (PDZ) domain, which initiates and/or facilitates the polymerization of claudins and is crucial for the formation of TJs [20]. These diverse interactions determine that ZO-1 has a scaffolding function. The JAMs are glycoproteins belonging to the immunoglobulin supergene family, which consists of two extracellular immunoglobulin-like structures, a transmembrane domain, and an intracellular region with PDZ binding sequence. The JAMs have multiple functions including the regulation of intestinal epithelial paracellular permeability [24, 25].

Characteristics of intestinal barrier dysfunction in severe burn injury

A large number of animal and clinical studies have demonstrated that the intestinal mechanical barrier is often disrupted by severe burn injury [26,27,28,29]. The reason for intestinal mechanical barrier dysfunction induced by severe burn injury is intricate and complex. Many factors such as neuroendocrine mediators, hypoxia/ischemia, complement activation, oxygen free radicals, inflammatory mediators, proinflammatory cytokines, and other mediators released in the stress response are directly or indirectly involved in the occurrence and development of intestinal mechanical barrier dysfunction associated with severe burn injury (Fig. 1). In addition, the disruption of intestinal mechanical barrier is also closely associated with burn shock, inflammation, infection, immune disturbance, hypermetabolism, sepsis, mutiple organ dysfunction syndrome, etc. [30, 31].

Fig. 1
figure 1

The schematic diagram illustrating the mechanisms of intestinal barrier dysfunction in severe burn injury. IFN-γ interferon-γ, TNF-α tumor necrosis factor, ILs interleukins, MLCK myosin light chain kinase, ROCK rho-associated protein kinase, MLC myosin light chain, TJPs tight junction proteins, ZO-1 zonula occludens

It has already been reported that the intestinal permeability is increased quickly and significantly in both patients and animals suffering from severe burn injury [32, 33]. Once the intestinal permeability is increased, the luminal bacteria and/or endotoxins translocate across the disrupted intestinal barrier, and then rapidly spread to distant organs such as the liver, spleen, lungs, and even the blood via the portal vein and/or the lymphatic system. The endotoxin translocation can arise as early as 15 min post-burn and reach the peak within 6 h [34]. Our previous studies have shown that in mice subjected to a 30% total body surface area (TBSA) full-thickness burn, the ileal permeability is markedly increased at 1 h, peaks at 6 h, and still significantly higher than control level at 24 h post-burn [26, 27, 35]. Similarly, in mice suffering from 30% TBSA full-thickness scald, the intestinal permeability is significantly increased at 2 h and reaches the peak at approximately 4–6 h, whereas the histological structure of intestinal mucosa is not changed at 2 h after injury [36, 37]. Thus, it is worthy to note that after severe burn injury, the increase of intestinal permeability does not synchronize with the damage of mucosal histological structure although the increased intestinal permeability is closely associated to the changes of mucosal histological structure. The intestinal hyperpermeability induced by severe burn injury can arise earlier than the mucosal histological changes. This phenomenon may be mainly attributed to the dysfunction of intestinal epithelial TJ barrier, but the impairment of mucosal histological structure definitely aggravates the increased permeability.

Factors contributing to the burn-induced intestinal barrier dysfunction

Stress

After severe burn injury, a variety of stressors such as mental stimulation, local tissue damage, ischemia/hypoxia, inflammation, and surgical operation can enhance the activities of both hypothalamic-pituitary-adrenal axis and sympathetic nerve system, leading to severe systemic stress responses. The stress may play an important role in the intestinal barrier dysfunction and hyperpermeability in some diseases including burn injury [38, 39]. An animal experiment has demonstrated the intestinal permeability, bacterial translocation, and proinflammatory cytokines such as interferon-γ (IFN-γ) are significantly increased by stress stimulation in C57BL/6 J mice, but not in the severe combined immunodeficiency (SCID) and IFN-γ-deficient mice [40], indicating that the stress-induced intestinal barrier dysfunction depends on the presence of CD4 + T lymphocytes and IFN-γ. In addition, inhibiting the phosphorylation of MLC by MLCK inhibitor ML-7 can significantly alleviate the stress-induced intestinal barrier dysfunction, suggesting that the MLCK-mediated MLC phosphorylation is involved in the stress-induced intestinal barrier dysfunction. Similarly, other investigators have also shown that acute stress can decrease the mRNA expression of TJ proteins (TJPs) ZO-2 and occludin, which in turn leads to intestinal barrier dysfunction [41].

Ischemia/hypoxia and ischemia-reperfusion injury

Extensive burn injuries cause extravasation of plasma, and then lead to hypovolemic shock, resulting in the ischemia and hypoxia of tissues. It is well recognized that ischemia/hypoxia is the most fundamental reason for systemic damages in the early stage of severe burn injury. Thus, ischemia/hypoxia is believed to play an important role in the pathogenesis of intestinal barrier dysfunction induced by severe burn injury. Previous studies have revealed that systemic or intestinal ischemia/reperfusion causes the intestinal barrier disruption, resulting in the increase of paracellular permeability [42,43,44]. It has been reported that the intestinal permeability is significantly increased 30 min after hemorrhagic shock, and the luminal bacteria translocate to the mesenteric lymph nodes, liver, and spleen [45]. The intestinal barrier disruption induced by hemorrhagic shock is accompanied by the activation of actin depolymerizing factor/cofilin, the increase of G-actin, and the decreases of F-actin, ZO-1, and claudin-3 in intestinal epithelia. Our previous study has shown that in mice subjected to 30% TBSA full-thickness flame burn, the intestinal permeability is significantly increased during the shock stage of burn and simultaneously accompanied by the decrease and redistribution of ZO-1 and the rearrangement of cytoskeleton F-actin [26]. Similarly, the intestinal permeability is significantly increased in severely scalded mice and also accompanied by the decreased expression and redistribution of both ZO-1 and occludin [35, 46]. In addition, our previous in vitro study has revealed that hypoxia alone can increase the permeability of cultured intestinal epithelial cell monolayers and cause the morphological changes of ZO-1 [47]. Thus, ischemia and/or hypoxia arising in the early stage of severe burn injury causes intestinal barrier dysfunction by disrupting the epithelial TJ, leading to the increase of permeability.

It has been well recognized that reperfusion after ischemia can produce large amounts of free radicals. The free radicals, on the one hand, can directly interact with the polyunsaturated fatty acid of the phospholipid membrane to form lipid peroxides, causing the impairment of intestinal epithelial cells. They, on the other hand, can also activate complements, mediate the release of inflammatory mediators, and induce cell apoptosis, leading to the disruption of the intestinal epithelial barrier. The free radicals have been demonstrated to induce the depolymerization and rearrangement of cytoskeleton F-actin and disrupt the intestinal epithelial barrier, in which PKC-λ activation is involved [48]. Moreover, reperfusion following ischemia not only damage the structure of intestinal epithelial TJ, but also alter the expression and localization of TJPs. Animal studies have shown that at 1 h post-reperfusion, the intestinal permeability is increased significantly while the histological structure of mucosa is obviously damaged as evidenced by the appearance of denuded villi, focal necrosis, ulcer, and bleeding. Meanwhile, the intestinal epithelial TJ is disrupted, and the expression and localization of ZO-1, occludin, and claudin-1, claudin-2, claudin-3, claudin-4, and claudin-5 are remarkedly changed following intestinal ischemia/reperfusion injury [49, 50].

Proinflammatory cytokines

After severe burn injury, large amounts of proinflammatory cytokines are released from both gastrointestinal tract and other organs [51]. Among these proinflammatory cytokines, IFN-γ, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 are believed to be the most important in inducing the intestinal epithelial barrier dysfunction and leading to increased intestinal permeability.

IFN-γ

Many previous in vivo and in vitro studies have confirmed that IFN-γ alone or in combination with other cytokines can induce intestinal epithelial barrier dysfunction, which is mainly associated with the altered expression and/or localization of TJPs in intestinal epithelial cells. IFN-γ has been reported to activate Rho-GTPase, upregulate the expression of Rho-associated protein kinase (ROCK), and induce the endocytosis of TJPs and the relocalization of occludin, claudin-1, and JAM-A via the RhoA/ROCK pathway, resulting in the intestinal epithelial barrier dysfunction [52]. Our in vitro studies have demonstrated that by inducing the expression of TNF receptor 2 (TNFR2) in the intestinal epithelial cells, IFN-γ synergizes with TNF-α to induce the expression of MLCK protein and to activate the MLCK-MLC phosphorylation pathway, which in turn results in the relocalization of TJPs ZO-1, occludin, and claudin-1, leading to the disruption of intestinal epithelial barrier function [53]. In addition, IFN-γ also downregulates the expressions of both ZO-1 and occludin by activating adenosine monophosphate-activated protein kinase, resulting in intestinal epithelial barrier dysfunction [54]. Moreover, IFN-γ induces the intestinal barrier dysfunction in vitro as assessed by the drop of transepithelial electrical resistance and the increases of both permeability and Escherichia coli transcytosis. However, inhibiting phosphatidylinositol 3′-kinase (PI-3 K) activity with LY294002 and wortmannin, the PI-3 K inhibitors, completely blocks the intestinal barrier dysfunction evoked by IFN-γ, suggesting the involvement of PI-3 K in the IFN-γ-induced intestinal barrier dysfunction [55]. Accordingly, IFN-γ disrupts intestinal epithelial barrier function via multiple signal transduction pathways.

TNF-α

A large number of previous studies have documented that TNF-α, alone or with other proinflammatory cytokines, can induce intestinal epithelial barrier disruption both in vivo and in vitro [56,57,58,59]. The underlying mechanisms involved in the TNF-α-induced intestinal epithelial barrier dysfunction include the apoptosis of intestinal epithelial cells, changes of the lipid composition in cell membrane, activation of MLCK by Ca2+-calmodulin, induction of MLC phosphorylation by upregulated MLCK protein expression, and inhibition of TJP expression, among which the MLCK-mediated MLC phosphorylation pathway is considered to play a critical role in the TNF-α-induced intestinal epithelial barrier dysfunction. It has been revealed that upon activated by TNF-α, nuclear factor of activated B cells protein kinase (NF-κB) binds to the NF-κB motif in the MLCK gene promoter, which initiates the MLCK gene transcription and then upregulates the MLCK protein expression, subsequently leading to intestinal epithelial barrier dysfunction [60]. Our previous in vitro studies have demonstrated that TNF-α acts synergistically with IFN-γ to induce intestinal epithelial barrier dysfunction in a dose-dependent manner, which is mediated by TNFR2 but not TNFR1. The molecular mechanism involves the activation of activator protein-1, which induces MLCK gene transcription, the upregulation of MLCK protein expression and MLC phosphorylation, and the relocalization of ZO-1, occludin, and claudin-1, resulting in the disrupted intestinal epithelial barrier [53, 61, 62]. In addition, our recent in vitro studies have demonstrated that hypoxia-inducible factor-1α (HIF-1α) is involved in the intestinal epithelial barrier dysfunction, the alteration of TJPs, and the increase of permeability induced by the synergistic action of TNF-α and IFN-γ, as evidenced by that the TNF-α and IFN-γ-induced intestinal epithelial barrier dysfunction is alleviated by both 3-(5'-hydroxymethyl-2'-furyl)-1-benzylind azole (YC)-1 and oligomycin, the specific chemical inhibitors of HIF-1α [63, 64]. We have also shown that lymphotoxin-like inducible protein that competes with glycoprotein D for herpes virus entry on T cells (LIGHT), a superfamily member of TNF (TNFSF14), disrupts the intestinal epithelial barrier by activating the MLCK-MLC phosphorylation pathway, which is mediated by the lymphotoxin β receptor (LTβR) belonging to the TNF receptor superfamily and the caveolin-1-dependent endocytosis of occludin [65]. The caveolin-1-dependent endocytosis of occludin is reported to be very important in the TNF-α-induced alteration of TJ [57].

Interleukins

Among the numerous ILs, IL-1β is the most studied in the regulation of intestinal epithelial barrier function. IL-1β has been well documented to be able to disrupt the intestinal epithelial barrier, leading to increased permeability [66,67,68,69]. The intestinal barrier disruption induced by IL-1β is most likely associated with the decreased expression and the relocalization of TJP occludin. The upregulated MLCK gene transcription triggered by NF-κB activation is also part of the mechanism by which IL-1β disrupts the intestinal epithelial barrier. It has been reported that the IL-1β-evoked increase of intestinal epithelial permeability is mediated by activation of extracellular signal-regulated kinases 1/2 (ERK1/2) signaling pathway and that inhibition of ERK1/2 activity inhibits the IL-1β-induced increase in intestinal epithelial permeability. The IL-1β-induced activation of ERK1/2 pathway leads to a downstream activation of nuclear transcription factor Elk-1. The activated Elk-1 translocates to the nucleus and binds to the cis-binding motif on MLCK promoter region, triggering MLCK gene activation, MLCK mRNA transcription, MLCK protein synthesis, and MLCK catalyzed opening of the intestinal epithelial TJ [70]. The p38 MAPK is also reported to be involved in the process of MLCK gene transcription induced by IL-1β. The IL-1β-induced increase of intestinal epithelial permeability is accompanied by the activation of p38 MAPK. The activated p38 MAPK then induces the phosphorylation and activation of activating transcription factor-2 (ATF-2), a substrate of p38 MAPK. The activated ATF-2 translocates to the nucleus where it attaches to its binding motif on the MLCK promoter region, leading to the activation of MLCK promoter activity and gene transcription. The IL-1β-induced activation of MLCK promoter and MLCK mRNA transcription is prevented by small interfering RNA induced silencing of ATF-2, or mutation of the ATF-2 binding motif on the MLCK promoter region [71]. Therefore, multiple signal transduction pathways are involved in the intestinal epithelial barrier dysfunction evoked by IL-1β, but the upregulation of MLCK gene transcription is the most critical.

Among the other ILs, IL-4 has been reported to disrupt intestinal barrier function via the PI-3 K signaling pathway [72], whereas the effect of IL-6 on intestinal barrier function is still controversial. On the one hand, it has been shown that IL-6 can induce intestinal barrier dysfunction and increase the permeability and that inhibition of IL-6 or knockout of the IL-6 gene can prevent the damage of intestinal mucosa and the increase of permeability in intestinal ischemia/reperfusion injury, sepsis, and hemorrhagic shock [73, 74]. An in vitro study has shown that IL-6 can only cause the increase of intestinal permeability to ions but not to large-molecule materials. This characteristic of IL-6 on the intestinal barrier is associated with the induced expression of claudin-2, but not ZO-1, ZO-2, occludin, JAM-1, or claudin-1, claudin-3, and claudin-4, in which the MEK/ERK and PI-3 K/Akt signal pathways are involved [75]. On the other hand, however, IL-6 has been reported to protect intestinal epithelial barrier function, and the potential mechanism may be related to the IL-6-induced upregulation of keratin-8 and 18 in intestinal epithelial cells [76]. Similarly, an in vivo study has demonstrated that IL-6 neutralizing antibody treatment can significantly reduce the ileal mucosa damage, bacterial translocation to the mesenteric lymph node, and relocalization of ZO-1 and occludin following the combined insult of ethanol exposure and burn injury; however, IL-6 gene knockout has no similar effects [77]. These findings suggest that maintaining the appropriate amount of IL-6 may be helpful to restore intestinal barrier function in inflammation, whereas complete loss of IL-6 seems not to be beneficial to maintain the intestinal barrier. In addition, IL-13 has also been reported to play an important role in the pathogenesis of intestinal barrier dysfunction. A recent study has shown that IL-13 induces the increased permeability of intestinal epithelium to ions in a dose-dependent manner, in which the IL-13-induced increase and redistribution of claudin-2 via signal transducers and activators of transcription 6 (STAT6) signaling pathway is involved [78].

Bacteria and endotoxins

Severe burn injury has been reported to be able to alter intestinal microbiota composition in both animals and patients, resulting in the intestinal microbiota dysbiosis [79,80,81]. The intestinal flora disturbance may increase the conditional pathogenic bacteria. Among the bacteria, enteropathogenic Escherichia coli (EPEC) is the most studied in the regulation of intestinal epithelial barrier function. Previous studies have determined that EPEC is capable of inducing the intestinal epithelial barrier dysfunction [82,83,84]. Utilizing its type III secretion system (T3SS), EPEC injects pathogenic effector proteins such as EspF, EspG, EspH, and Map into the intestinal epithelial cells, causing the cytoskeleton collapse and relocalization of TJPs. The resulting compromised barrier and increased intestinal permeability may be responsible for the clinical symptoms of EPEC infection. Some in vivo studies have revealed that EPEC infection causes the disruption of the intestinal barrier and the relocalization of TJPs. The disruptive effect of EPEC on the intestinal epithelial barrier is dependent on the pathogenic effector protein EspF [85,86,87]. Notably, the EPEC-induced intestinal barrier dysfunction is also related to the activation of the MLCK-MLC phosphorylation pathway, because that inhibition of MLCK with ML-9, a specific chemical inhibitor of MLCK, can prevent the intestinal barrier defects induced by EPEC infection [88]. The PKCε signaling pathway is also reported to be involved in the intestinal epithelial barrier disruption induced by EPEC infection [89]. In addition, enteroinvasive Escherichia coli (EIEC) has also been demonstrated to evoke the relocalization and reduced expression of ZO-1, occludin, claudin-1, and JAM-1 in the intestinal epithelial cells, resulting in the intestinal epithelial barrier dysfunction [90].

A huge number of studies have determined the disruptive effect of endotoxin on intestinal epithelial barrier both in vivo and in vitro. On the one hand, endotoxin disrupts the intestinal barrier by inducing a variety of cells to produce proinflammatory mediators such as TNF-α and IL-1β. On the other hand, endotoxin also directly impairs intestinal epithelial cells to cause the barrier dysfunction. An in vivo study has shown that intestinal permeability and bacterial translocation is significantly increased and the intestinal epithelial TJs are obviously opened in lipopolysaccharide (LPS)-challenged rats. This disruptive effect of LPS on intestinal barrier depends on MLCK, because that inhibiting MLCK activity with ML-7, a specific inhibitor of MLCK, can alleviate the intestinal barrier dysfunction and bacterial translocation in rats challenged with LPS [91]. Moreover, LPS has been reported to cause the relocalization of ZO-1, occludin, and claudin-1 and claudin-4 and reduce the expression of ZO-1, in which c-Src, toll-like receptor 4 (TLR4), and LPS binding protein are involved [92]. Our latest in vitro study has revealed that LPS also induces epithelial barrier disruption by activating Nod-like receptor protein 3 (NLRP3) inflammasome and autophagy in Caco-2 intestinal epithelial monolayers [93].

Mechanisms involved in the burn-induced intestinal barrier dysfunction

Alteration of TJPs

It is well known that the structural basis of intestinal epithelial barrier integrity is TJ and its associated proteins such as ZOs, occludin, and claudins. However, the changes in intestinal epithelial TJPs after severe burn injury are not well defined so far. In general, there are mainly two aspects about the changes in intestinal epithelial TJPs after severe burns: one is the relocalization or redistribution of the TJPs in intestinal epithelial cells, and the other is the altered expression of TJPs. Our previous study has demonstrated that there is an obvious relocalization of ZO-1, occludin, and claudin-1 in the ileal mucosa of mice inflicted with 30% TBSA full-thickness flame burns [94]. Other investigators have also shown that the protein expression of occludin in intestinal epithelial cells is not significantly changed in rats subjected to a 30% TBSA burn, but the relocalization of occludin is evident after burn injury. There is a clearly decreased expression and relocalization of occludin in the ileum of burned rats combined with Enterococcus faecalis infection [95]. It has been reported that the expressions of ZO-1 and occludin in intestinal epithelia are decreased to the lowest at 6 h post-burn, with reductions of 68% and 43% respectively, and still significantly lower than control level at 24 h post-burn [46]. Furthermore, simple hemorrhagic shock can also induce the decreased expression and relocalization of ZO-1 and claudin-3, leading to intestinal TJ barrier dysfunction and bacterial translocation [45]. Therefore, both the decrease and relocalization of TJPs in intestinal epithelial cells after severe burn injury are bound to impair the structure and function of intestinal epithelial TJ, resulting in barrier disruption and hyperpermeability.

Role of MLC phosphorylation

It is well established that the TJs are connected to cytoskeleton actin through ZOs [22, 96,97,98]. Thus, the integrity of the intestinal TJ barrier also depends on the structural assembly and functional status of the intestinal epithelial cytoskeleton actin as well as the interaction between myosin and actin. A critical step in the regulation of epithelial TJ permeability may be myosin ATPase-mediated contraction of the perijunctional actomyosin ring and subsequent physical tension on the TJ [99], to which MLC phosphorylation is critical. It is well known that MLC phosphorylation is regulated by both MLCK and MLC phosphatase (MLCP) [100], which is more commonly called myosin phosphatase. On the one hand, MLCK phosphorylates MLC, resulting in an increase of MLC phosphorylation. On the other hand, MLCP dephosphorylates MLC, leading to a reduction of MLC phosphorylation. It has been reported that the induction of MLC phosphorylation in intestinal epithelial cells alone is sufficient to change the TJPs ZO-1 and occludin, leading to the increased permeability of the intestinal epithelial TJ [101]. Our previous animal studies have shown that the remarkably increased intestinal permeability and the redistribution of TJPs following severe burn injury are accompanied by the significantly increased MLC phosphorylation [26, 27, 94]. Similarly, our in vitro studies have also revealed that the barrier disruption caused by burn sera or hypoxia is accompanied by a significant increase of MLC phosphorylation in cultured intestinal epithelial cell monolayers [47, 94]. Thus, the increased MLC phosphorylation may be a central molecular mechanism involved in the severe burn-induced intestinal barrier disruption and hyperpermeability.

Role of MLCK

It is well known that MLCK, a member of the serine/threonine protein kinase family, is a Ca2+/calmodulin-dependent kinase responsible for the phosphorylation of a specific serine in the N-terminus of MLC. Upon activated by upstream signals such as Ca2+, histamine, bradykinin, free radicals, and proinflammatory cytokines, MLCK can induce the phosphorylation of serine 18/threonine 19 of MLC in nonmuscle cells. It has been well documented that MLCK is a central determinant in the intestinal epithelial barrier dysfunction under various pathological conditions, and the underlying mechanism involves the induction of MLC phosphorylation. Based on the reported studies, it is believed that MLCK also plays a critical role in intestinal barrier dysfunction after severe burn injury. Some previous studies have shown that MLCK is critical to intestinal epithelial barrier dysfunction, increased permeability, and the relocalization of TJPs ZO-1, occludin, and claudins induced by a number of pathophysiological conditions associated with severe burns such as stress, shock, ischemia/hypoxia, inflammation, and infection [5, 39, 47, 97, 101,102,103,104]. The previous animal studies have revealed that MLCK chemical inhibitor ML-9 or peptide inhibitor PIK could significantly alleviate the barrier dysfunction, the changes of TJPs, the increase of MLC phosphorylation, and the hyperpermeability of intestinal epithelia in mice subjected to severe burns [94, 105]. Similarly, in MLCK knockout mice inflicted with 40% TBSA full-thickness burn, there was only slight elevation of intestinal permeability at 4 h after the injury, without obvious damage to the intestinal mucosa [106]. Therefore, after severe burn injury, MLCK upregulates the phosphorylation of MLC in intestinal epithelial cells, thereby leading to the barrier dysfunction and hyperpermeability.

Role of ROCK

Rho, which belongs to the Ras superfamily of low molecular weight GTPases, is also an important regulator of the intestinal epithelial barrier. It regulates the permeability of the intestinal barrier by mainly affecting the interaction between actin and myosin in epithelial cells. The downstream effector of RhoA is ROCK, a serine/threonine protein kinase. The activated ROCK phosphorylates the myosin phosphatase target subunit (MYPT), which is also called the myosin-binding subunit (MBS) of myosin phosphatase, and consequently inactivates myosin phosphatase, thereby increasing the phosphorylation of MLC. In other words, the activation of ROCK results in an increase of MLC phosphorylation. At present, there are few data about the involvement of ROCK in post-burn intestinal barrier dysfunction. However, some studies have determined that ROCK is involved in the intestinal epithelial barrier disruption caused by proinflammatory cytokines, inflammation, and bacteria [52, 107, 108]. For example, ROCK has been reported to mediate the bacteria-induced increase of intestinal epithelial permeability and the decreased expression and relocalization of claudin-4 and claudin-5 [108]. Our previous study has demonstrated that intestinal permeability, ROCK protein expression, and MLC phosphorylation are elevated in mice subjected to 30% TBSA full-thickness burns. However, intraperitoneal injection of ROCK-specific inhibitor Y-27632 immediately after the injury can significantly alleviate the severe burn-induced increases of intestinal permeability and MLC phosphorylation [27]. Similarly, burn sera can induce barrier dysfunction and upregulate ROCK protein expression and MLC phosphorylation in cultured intestinal epithelial cell monolayers, whereas inhibiting ROCK activity with Y-27632 can also alleviate the burn sera-induced barrier dysfunction and the increase of MLC phosphorylation in cultured intestinal epithelial cell monolayers [109]. Therefore, ROCK is believed to be involved in the severe burn-induced intestinal barrier dysfunction and hyperpermeability by inducing MLC phosphorylation.

Therapeutic strategies for the burn-induced intestinal barrier dysfunction

It is well recognized that the treatment of post-burn intestinal barrier dysfunction is an important part of the burn treatment and directly related to the level of comprehensive treatment of severe burns. The factors or pathophysiological processes affecting intestinal barrier function after severe burns are multiple and complex. Thus, the appropriate therapeutic measures should correspondingly be taken to every critical pathophysiological process, such as stress, shock, ischemia/hypoxia, inflammation, infection, and surgical operation. Many clinical and experimental studies in the past have suggested that taking some positive and reasonable measures is beneficial to the intestinal barrier in the early stage of severe burns. These measures include positive anti-shock to improve the oxygen supply to organs including intestine, control of inflammation and infection, wound management, early enteral nutrition, immunonutrition, ecoimmunonutrition, and supplementation of some special nutrients such as glutamine and arginine [2, 30, 31].

In addition to the aforementioned therapeutic measures, an animal study has recently demonstrated that zinc finger aspartate-histidine-histidine-cysteine (DHHC) domain-containing protein-21 (ZDHHC21), a particular palmitoyl acyltransferase, mediates the intestinal epithelial hyperpermeability in mice subjected to a 40% TBSA full-thickness scald injury [110]. The thermal injury-induced intestinal barrier dysfunction is significantly attenuated in mice with genetic ablation of ZDHHC21 or by intraperitoneal injection of 2-bromopalmitate, a pharmacological inhibitor of palmitoyl acyltransferases, suggesting that targeting palmitoyl acyltransferase ZDHHC21 can effectively attenuate the intestinal epithelial barrier disruption caused by severe burn injury. Moreover, endoplasmic reticulum stress (ERS)-autophagy pathway has recently been demonstrated to be associated with intestinal epithelial TJ barrier dysfunction induced by severe burn injury [35]. Inhibiting ERS or autophagy with specific inhibitor can significantly ameliorate the severe burn-induced intestinal TJ barrier dysfunction [111]. It is implied that blocking ERS-autophagy pathway may be beneficial to restoring intestinal epithelial TJ barrier dysfunction induced by severe burn injury. Furthermore, it is very interesting that mesalazine, which is also known as 5-aminosalicylic acid and a common clinical anti-inflammatory drug used to treat inflammatory bowel disease, has recently been reported to be able to treat the intestinal barrier disruption resulting from burn injury [112]. Treatment with mesalazine after burn injury prevents the burn-induced increase of intestinal permeability, normalizes the levels of pro-inflammatory cytokines, and restores the expression of TJPs claudin-4 and occludin, which indicates that mesalazine can potentially be used as the therapeutic drug for intestinal barrier disruption induced by severe burn injury.

With the deepening understanding of the regulatory mechanisms of intestinal epithelial TJ, new therapeutic strategies aiming at the regulation of intestinal epithelial TJ may be the direction for the prevention and treatment of post-burn intestinal barrier dysfunction, especially the measures targeting at MLC phosphorylation. This may be more important for controlling increased permeability when the histological structure of intestinal mucosa has not yet been damaged obviously. The previous animal studies have already demonstrated that inhibiting MLCK and ROCK activity by using specific inhibitors to reduce MLC phosphorylation in intestinal epithelial cells can significantly alleviate the severe burn-induced intestinal barrier dysfunction and hyperpermeability [27, 94, 105]. Moreover, it has been reported that both phosphodiesterase inhibitor pentoxifylline and p38 MAPK inhibitor SB203580 can reduce intestinal permeability and protect intestinal barrier function in severe burn injury, the underlying mechanism of which is proved to be mediated by inhibiting the MLCK-mediated upregulation of MLC phosphorylation [36, 113]. Another in vivo study has also revealed that inhibiting MLCK-mediated MLC phosphorylation by MLCK inhibitor ML-7 can prevent the increase of intestinal permeability, opening of TJ, bacterial translocation, and intestinal inflammatory response in rats challenged with endotoxin [91]. Most notably, the latest study has revealed that divertin, a domain-binding small molecule that blocks MLCK1 recruitment without inhibiting enzymatic function, is capable of blocking acute, TNF-induced MLCK1 recruitment as well as downstream MLC phosphorylation, barrier loss, and diarrhea both in vitro and in vivo, suggesting that MLCK1 diversion can reverse intestinal epithelial barrier loss [114]. Thus, although there are still many issues that require to be further clarified, such as the effectiveness, safety, and feasibility on clinical applications, the therapeutic strategies targeting at MLC phosphorylation pathway may be the direction of future efforts in the prevention and treatment of intestinal barrier dysfunction after severe burn injury.

Conclusions

The integrity of intestinal barrier function is essential to maintain the homeostasis of the intestinal mucosa. However, the intestinal barrier is disrupted after severe burn injury. A broad range of pathogenic factors is known to induce the disruption of epithelial TJ barrier, thereby leading to the intestinal hyperpermeability. The mechanism of intestinal barrier disruption induced by severe burn injury is extremely complex, involving numerous signaling molecules and related pathways. A better basic understanding of the mechanism might be helpful for the prevention or treatment of intestinal barrier disruption following severe burn injury.

Availability of data and materials

Not applicable

Abbreviations

Elk-1:

Ets-like gene 1

EPEC:

Enteropathogenic Escherichia coli

ERK:

Extracellular signal-regulated kinase

ERS:

Endoplasmic reticulum stress

IFN-γ:

Interferon-γ

IL:

Interleukin

JAM:

Junctional adhesion molecule

LIGHT:

Lymphotoxin-like inducible protein that competes with glycoprotein D for herpes virus entry on T cells

LPS:

Lipopolysaccharide

MAPK:

Mitogen-activated protein kinases

MLC:

Myosin light chain

MLCK:

Myosin light chain kinase

NLRP3:

Nod-like receptor protein 3

PDZ:

Postsynaptic density protein-95/Discs-Large/zonula occludens-1

PI-3 K:

Phosphatidylinositol 3′-kinase

PKC:

Protein kinase C

ROCK:

Rho-associated protein kinase

SCID:

Severe combined immunodeficiency

STAT6:

Signal transducers and activators of transcription 6

TBSA:

Total body surface area

TJ:

Tight junction

TJP:

Tight junction protein

TLR4:

Toll-like receptor 4

TNFR:

Tumor necrosis factor receptor

TNF-α:

Tumor necrosis factor-α

ZDHHC21:

Zinc finger aspartate-histidine-histidine-cysteine domain-containing protein-21

ZO:

Zonula occludens

References

  1. Ziegler TR, Smith RJ, O’Dwyer ST, Demling RH, Wilmore DW. Increased intestinal permeability associated with infection in burn patients. Arch Surg. 1988;123:1313–9.

    Article  CAS  PubMed  Google Scholar 

  2. De-Souza DA, Greene LJ. Intestinal permeability and systemic infections in critically ill patients: effect of glutamine. Crit Care Med. 2005;33:1125–35.

    Article  PubMed  Google Scholar 

  3. Al-Ghoul WM, Khan M, Fazal N, Sayeed MM. Mechanisms of postburn intestinal barrier dysfunction in the rat: roles of epithelial cell renewal, E-cadherin, and neutrophil extravasation. Crit Care Med. 2004;32:1730–9.

    Article  CAS  PubMed  Google Scholar 

  4. Kraehenbuhl JP, Pringault E, Neutra M. Intestinal epithelia and barrier functions. Aliment Pharmacol Ther. 1997;11(3):3–8.

    PubMed  Google Scholar 

  5. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9(11):799–809.

    Article  CAS  PubMed  Google Scholar 

  6. Marchiando AM, Graham WV, Turner JR. Epithelial barriers in homeostasis and disease. Annu Rev Pathol-Mech. 2010;5:119–44.

    Article  CAS  Google Scholar 

  7. Cecilia Berin M, Li H, Sperber K. Antibody-mediated antigen sampling across intestinal epithelial barriers. Ann N Y Acad Sci. 2006;1072:253–61.

    Article  PubMed  CAS  Google Scholar 

  8. Fink MP. Intestinal epithelial hyperpermeability: update on the pathogenesis of gut mucosal barrier dysfunction in critical illness. Curr Opin Crit Care. 2003;9(2):143–51.

    Article  PubMed  Google Scholar 

  9. Gonzalez-Mariscal L, Betanzos A, Nava P, Jaramillo BE. Tight junction proteins. Prog Biophys Mol Biol. 2003;81:1–44.

    Article  CAS  PubMed  Google Scholar 

  10. Cereijido M, Contreras RG, Flores-Bentez D, Flores-Maldonado C, Larre I, Ruiz A, et al. New diseases derived or associated with the tight junction. Arch Med Res. 2007;38:465–78.

    Article  CAS  PubMed  Google Scholar 

  11. Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junction pore and leak pathways: a dynamic duo. Annu Rev Physiol. 2011;73:283–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S, Tsukita S. Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol. 1993;123(6):1777–88.

    Article  CAS  PubMed  Google Scholar 

  13. Schneeberger EE, Lynch RD. The tight junction: a multifunctional complex. Am J Physiol Cell Physiol. 2004;286(6):C1213–28.

    Article  CAS  PubMed  Google Scholar 

  14. Feldman GJ, Mullin JM, Ryan MP. Occludin: structure, function and regulation. Adv Drug Deliv Rev. 2005;57(6):883–917.

    Article  CAS  PubMed  Google Scholar 

  15. Buschmann MM, Shen L, Rajapakse H, Raleigh DR, Wang Y, Wang Y, et al. Occludin OCEL domain interactions are required for maintenance and regulation of the tight junction barrier to macromolecular flux. Mol Biol Cell. 2013;24(19):3056–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Van Itallie CM, Anderson JM. Claudins and epithelial paracellular transport. Rev Physiol. 2006;68(1):403–29.

    Article  CAS  Google Scholar 

  17. Bücker R, Schumann M, Amasheh S, Schulzke JD. Claudins in intestinal function and disease. Curr Top Membr. 2010;65:195–227.

    Article  CAS  Google Scholar 

  18. Lu Z, Ding L, Lu Q, Chen YH. Claudins in intestines: distribution and functional significance in health and diseases. Tissue Barriers. 2013;1(3):e24978.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Smalley KS, Brafford P, Haass NK, Brander JM, Brown E, Herlyn M. Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am J Pathol. 2005;166(5):1541–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Umeda K, Ikenouchi J, Katahira-Tayama S, Furuse K, Sasaki H, Nakayama M, et al. ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell. 2006;126(4):741–54.

    Article  CAS  PubMed  Google Scholar 

  21. Shin K, Margolis B. ZOning out tight junctions. Cell. 2006;126(4):647–9.

    Article  CAS  PubMed  Google Scholar 

  22. Fasano A. Physiological, pathological, and therapeutic implications of zonulin-mediated intestinal barrier modulation: living life on the edge of the wall. Am J Pathol. 2008;173(5):1243–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Van Itallie CM, Fanning AS, Bridges A, Anderson JM. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol Biol Cell. 2009;20(17):3930–40.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bazzoni G. Pathobiology of junctional adhesion molecules. Antioxid Redox Signal. 2011;15:1221–34.

    Article  CAS  PubMed  Google Scholar 

  25. Luissint AC, Nusrat A, Parkos CA. JAM-related proteins in mucosal homeostasis and inflammation. Semin Immunopathol. 2014;36(2):211–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen CL, Liu YL, Wang P, Sun W, Wang FJ. Role of MLC phosphorylation in intestinal epithelial barrier dysfunction induced by severe burn injury. Acta Academ Med Milit Tert. 2008;30(15):1434–7.

    CAS  Google Scholar 

  27. Liu YL, Wang FJ, Chen CL, Wang P. Increased intestinal permeability in severely burnt rats: regulatory mechanism of Rho kinase. Acta Academ Med Milit Tert. 2008;30(9):817–9.

    CAS  Google Scholar 

  28. Peterson CY, Costantini TW, Loomis WH, Putnam JG, Wolf P, Bansal V, et al. Toll-like receptor-4 mediates intestinal barrier breakdown after thermal injury. Surg Infect. 2010;11(2):137–44.

    Article  Google Scholar 

  29. Yang XK, Chen J, Bai H, Tao K, Zhou Q, Hou HY, et al. Inhibition of Na+/H+ exchanger 1 by cariporide reduces burn-induced intestinal barrier breakdown. Burns. 2013;39(8):1557–64.

    Article  PubMed  Google Scholar 

  30. Wang FJ. Research on intestinal tight junction barrier dysfunction should be emphasized in burn injury. Chin J Burns. 2010;26(5):331–3.

    CAS  Google Scholar 

  31. Audra C, Imran J, Madni T, Wolf SE. Nutrition and metabolism in burn patients. Burns & Trauma. 2017;5:11.

    Article  Google Scholar 

  32. Deitch EA. Intestinal permeability is increased in burn patients shortly after injury. Surgery. 1990;77(107):411–6.

    Google Scholar 

  33. Epstein MD, Tchervenkov JI, Alexander JW, Johnson JR, Vester JW. Increased gut permeability following burn trauma. Arch Surg. 1991;126(2):198–200.

    Article  CAS  PubMed  Google Scholar 

  34. Xiao GX. The gut-origin infection in severe bums. Chin J Burns. 2008;24(5):331–3.

    Google Scholar 

  35. Huang YL, Feng YH, Wang Y, Wang P, Wang FJ, Ren H. Severe burn-induced intestinal epithelial barrier dysfunction is associated with endoplasmic reticulum stress and autophagy in mice. Front Physiol. 2018;9:441.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Costantini TW, Loomis WH, Putnam JG, Kroll L, Eliceiri BP, Baird A, et al. Pentoxifylline modulates intestinal tight junction signaling after burn injury: effects on myosin light chain kinase. J Trauma. 2009;66(1):17–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Costantini TW, Eliceiri BP, Peterson CY, Loomis WH, Putnam JG, Baird A, et al. Quantitative assessment of intestinal injury using a novel in vivo, near-infrared imaging technique. Mol Imaging. 2010;9(1):30–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Collins SM. Stress and the gastrointestinal tract IV. Modulation of intestinal inflammation by stress: basic mechanisms and clinical relevance. Am J Phys. 2001;280:G315–8.

    CAS  Google Scholar 

  39. Buret AG. How stress induces intestinal hypersensitivity. Am J Pathol. 2006;168(1):3–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ferrier L, Mazelin L, Cenac N, Desreumaux P, Janin A, Emilie D, et al. Stress-induced disruption of colonic epithelial barrier: role of interferon-γ and myosin light chain kinase in mice. Gastroenterology. 2003;125(3):795–804.

    Article  CAS  PubMed  Google Scholar 

  41. Demaude J, Salvador-Cartier C, Fioramonti J, Ferrier L, Bueno L. Phenotypic changes in colonocytes following acute stress or activation of mast cells in mice: implications for delayed epithelial barrier dysfunction. Gut. 2006;55(5):655–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sun Z, Wang X, Deng X, Lasson A, Wallén R, Hallberg E, et al. The influence of intestinal ischemia and reperfusion on bidirectional intestinal barrier permeability, cellular membrane integrity, proteinase inhibitors, and cell death in rats. Shock. 1998;10(3):203–12.

    Article  CAS  PubMed  Google Scholar 

  43. Wattanasirichaigoon S, Menconi MJ, Delude RL, Fink MP. Effect of mesenteric ischemia and reperfusion or hemorrhagic shock on intestinal mucosal permeability and ATP content in rats. Shock. 1999;12(2):127–33.

    Article  CAS  PubMed  Google Scholar 

  44. Drewe J, Beglinger C, Fricker G. Effect of ischemia on intestinal permeability of lipopolysaccharides. Eur J Clin Investig. 2001;31(2):138–44.

    Article  CAS  Google Scholar 

  45. Thuijls G, de Haan JJ, Derikx JP, Daissormont I, Hadfoune M, Heineman E, et al. Intestinal cytoskeleton degradation precedes tight junction loss following hemorrhagic shock. Shock. 2009;31(2):164–9.

    Article  CAS  PubMed  Google Scholar 

  46. Costantini TW, Loomis WH, Putnam JG, Drusinsky D, Deree J, Choi S, et al. Burn-induced gut barrier injury is attenuated by phosphodiesterase inhibition: effects on tight junction structural proteins. Shock. 2009;31(4):416–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang P, Chen CL, Li M, Wang FJ. The role myosin light chain kinase in intestinal barrier dysfunction due to hypoxia. Chin J Burns. 2009;25(1):57–60.

    Google Scholar 

  48. Banan A, Zhang LJ, Farhadi A, Fields JZ, Shaikh M, Forsyth CB, et al. Critical role of the atypical λ isoform of protein kinase C (PKC-λ) in oxidant-induced disruption of the microtubule cytoskeleton and barrier function of intestinal epithelium. J Pharmacol Exp Ther. 2005;312(2):458–71.

    Article  CAS  PubMed  Google Scholar 

  49. Inoue K, Oyamada M, Mitsufuji S, Okanoue T, Takamatsu T. Different changes in the expression of multiple kinds of tight-junction proteins during ischemia-reperfusion injury of the rat ileum. Acta Histochem Cytochem. 2006;39(2):35–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li Q, Zhang Q, Wang C, Liu X, Qu L, Gu L, et al. Altered distribution of tight junction proteins after intestinal ischaemia/reperfusion injury in rats. J Cell Mol Med. 2009;13(9B):4061–76.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Finnerty CC, Herndon DN, Chinkes DL, Jeschke MG. Serum cytokine differences in severely burned children with and without sepsis. Shock. 2007;27(1):4–9.

    Article  CAS  PubMed  Google Scholar 

  52. Utech M, Ivanov AI, Samarin SN, Bruewer M, Turner JR, Mrsny RJ, et al. Mechanism of IFN-γ-induced endocytosis of tight junction proteins: myosin II-dependent vacuolarization of the apical plasma membrane. Mol Biol Cell. 2005;16(10):5040–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang FJ, Schwarz BT, Graham WV, Wang Y, Su L, Clayburgh DR, et al. IFN-γ-induced TNFR2 up-regulation is required for TNF-dependent intestinal epithelial barrier dysfunction. Gastroenterology. 2006;131(4):1153–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Scharl M, Paul G, Barrett KE, McCole DF. Adenosine monophosphate activated protein kinase mediates the interferon gamma-induced decrease in intestinal epithelial barrier function. J Biol Chem. 2009;284(41):27952–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. McKay DM, Watson JL, Wang A, Caldwell J, Prescott D, Ceponis PMJ, et al. Phosphatidylinositol 3′-kinase is a critical mediator of interferon-γ-induced increases in enteric epithelial permeability. J Pharmacol Exp Ther. 2007;320(3):1013–22.

    Article  CAS  PubMed  Google Scholar 

  56. Clayburgh DR, Musch MW, Leitges M, Fu YX, Turner JR. Coordinated epithelial NHE3 inhibition and barrier dysfunction are required for TNF-mediated diarrhea in vivo. J Clin Invest. 2006;116(10):2682–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Marchiando AM, Shen L, Graham WV, Weber CR, Schwarz BT, Austin JR II, et al. Caveolin-1-dependent occludin endocytosis is required for TNF-induced tight junction regulation in vivo. J Cell Biol. 2010;189(1):111–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Fischer A, Gluth M, Pape UF, Wiedenmann B, Theuring F, Baumgart DC. Adalimumab prevents barrier dysfunction and antagonizes distinct effects of TNF on tight junction proteins and signaling pathways in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol. 2013;304(11):970–9.

    Article  CAS  Google Scholar 

  59. Feng Y, Teitelbaum DH. TNF-α-induced loss of intestinal barrier function requires TNFR1 and TNFR2 signaling in a mouse model of total parenteral nutrition. J Physiol. 2013;591(15):3709–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ye D, Ma TY. Cellular and molecular mechanisms that mediate basal and tumor necrosis factor-α induced regulation of myosin light chain kinase gene activity. J Cell Mol Med. 2008;12(4):1331–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang FJ, Graham WV, Wang YM, Witkowski ED, Schwarz BT, Turner JR. Interferon-γ and tumor necrosis factor-α synergize to induce intestinal epithelial barrier dysfunction by upregulating myosin II regulatory light chain kinase expression. Am J Pathol. 2005;166(2):409–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Graham WV, Wang FJ, Clayburgh DR, Cheng JX, Yoon B, Wang Y, et al. Tumor necrosis factor-induced long myosin light chain kinase transcription is regulated by differentiation-dependent signaling events: characterization of the human long myosin light chain kinase promoter. J Biol Chem. 2006;281(36):26205–15.

    Article  CAS  PubMed  Google Scholar 

  63. Liu H, Li M, Wang P, Wang FJ. Blockade of hypoxia-inducible factor-1α by YC-1 attenuates interferon-γ and tumor necrosis factor-α-induced intestinal epithelial barrier dysfunction. Cytokine. 2011;56(3):581–8.

    Article  CAS  PubMed  Google Scholar 

  64. Liu H, Wang P, Cao M, Li M, Wang FJ. Protective role of oligomycin against intestinal epithelial barrier dysfunction caused by IFN-γ and TNF-α. Cell Physiol Biochem. 2012;29(5-6):799–808.

    Article  CAS  PubMed  Google Scholar 

  65. Schwarz BT, Wang FJ, Shen L, Clayburgh DR, Su L, Wang Y, et al. LIGHT signals directly to intestinal epithelia to cause barrier dysfunction via cytoskeletal and endocytic mechanisms. Gastroenterology. 2007;132(7):2383–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Al-Sadi RM, Ma TY. IL-1β causes an increase in intestinal epithelial tight junction permeability. J Immunol. 2007;178(7):4641–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Al-Sadi R, Ye D, Dokladny K, Ma TY. Mechanism of IL-1β-induced increase in intestinal epithelial tight junction permeability. J Immunol. 2008;180(8):5653–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Al-Sadi R, Ye D, Said HM, Ma TY. IL-1β-induced increase in intestinal epithelial tight junction permeability is mediated by MEKK-1 activation of canonical NF-κB pathway. Am J Pathol. 2010;177(5):2310–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Al-Sadi R, Guo S, Dokladny K, Smith MA, Ye D, Kaza A, et al. Mechanism of interleukin-1β iInduced-increase in mouse intestinal permeability in vivo. J Interf Cytok Res. 2012;32(10):474–84.

    Article  CAS  Google Scholar 

  70. Al-Sadi R, Ye D, Said HM, Ma TY. Cellular and molecular mechanism of interleukin-1β modulation of CACO-2 intestinal epithelial tight junction barrier. J Cell Mol Med. 2011;15(4):970–82.

    Article  CAS  PubMed  Google Scholar 

  71. Al-Sadi R, Guo S, Ye D, Dokladny K, Alhmoud T, Ereifej L, et al. Mechanism of IL-1β modulation of intestinal epithelial barrier involves p38 kinase and activating transcription factor-2 activation. J Immunol. 2013;190(12):6596–606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Di Leo V, Yang PC, Berin MC, Perdue MH. Factors regulating the effect of IL-4 on intestinal epithelial barrier function. Int Arch Allergy Immunol. 2002;129(3):219–27.

    Article  PubMed  CAS  Google Scholar 

  73. Yang R, Han X, Uchiyama T, Watkins SK, Yaguchi A, Delude RL, et al. IL-6 is essential for development of gut barrier dysfunction after hemorrhagic shock and resuscitation in mice. Am J Physiol Gastrointest Liver Physiol. 2003;285(3):G621–9.

    Article  CAS  PubMed  Google Scholar 

  74. Wang Q, Fang CH, Hasselgren PO. Intestinal permeability is reduced and IL-10 levels are increased in septic IL-6 knockout mice. Am J Physiol Regul Integr Comp Physiol. 2001;281(3):R1013–23.

    Article  CAS  PubMed  Google Scholar 

  75. Suzuki T, Yoshinaga N, Tanabe S. IL-6 regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286(36):31263–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wang L, Srinivasan S, Theiss AL, Merlin D, Sitaraman SV. Interleukin-6 induces keratin expression in intestinal epithelial cells: potential role of keratin-8 in interleukin-6-induced barrier function alterations. J Biol Chem. 2007;282(11):8219–27.

    Article  CAS  PubMed  Google Scholar 

  77. Zahs A, Bird MD, Ramirez L, Choudhry MA, Kovacs EJ. Anti-IL-6 antibody treatment but not IL-6 knockout improves intestinal barrier function and reduces inflammation following binge ethanol exposure and burn injury. Shock. 2013;39(4):373–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Weber CR, Raleigh DR, Su L, Shen L, Sullivan EA, Wang Y, et al. Epithelial myosin light chain kinase activation induces interleukin-13 expression to alter tight junction ion selectivity. J Biol Chem. 2010;285(16):12037–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Shimizu K, Oqura H, Asahara T, Nomoko K, Matsushima A, Hayakawa K, et al. Gut microbiota and environment in patients with major burns-a preliminary report. Burns. 2015;41(3):e28–33.

    Article  PubMed  Google Scholar 

  80. Earley ZM, Akhtar S, Green SJ, Naqib A, Khan O, Cannon AR, et al. Burn injury alters the intestinal microbiome and increases gut permeability and bacterial translocation. PLoS One. 2015;10(7):e0129996.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Feng YH, Huang YL, Wang Y, Wang P, Wang FJ. Severe burn injury alters intestinal microbiota composition and impairs intestinal barrier in mice. Burns & Trauma. 2019;7:in press. https://doi.org/10.1186/s41038-019-0156-1.

  82. 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. 2001;107:621–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Guttman JA, Finlay BB. Tight junctions as targets of infectious agents. Biochim Biophys Acta. 2009;1788(4):832–41.

    Article  CAS  PubMed  Google Scholar 

  84. Long TM, Nisa S, Donnenberg MS, Hassel BA. Enteropathogenic Escherichia coli inhibits type I interferon- and RNase L-mediated host defense to disrupt intestinal epithelial cell barrier function. Infect Immun. 2014;82(7):2802–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Shifflett DE, Clayburgh DR, Koutsouris A, Turner JR, Hecht GA. Enteropathogenic E. coli disrupts tight junction barrier function and structure in vivo. Lab Investig. 2005;85:1308–24.

    Article  CAS  PubMed  Google Scholar 

  86. Zhang Q, Li Q, Wang C, Li N, Li J. Redistribution of tight junction proteins during EPEC infection in vivo. Inflammation. 2012;35:23–32.

    Article  PubMed  CAS  Google Scholar 

  87. Zhang Q, Li Q, Wang C, Liu X, Li N, Li J. Enteropathogenic Escherichia coli changes distribution of occludin and ZO-1 in tight junction membrane microdomains in vivo. Microb Pathog. 2010;48(1):28–34.

    Article  PubMed  CAS  Google Scholar 

  88. Yuhan R, Koutsouris A, Savkovic SD, Hecht G. Enteropathogenic Escherichia coli-induced myosin light chain phosphorylation alters intestinal epithelial permeability. Gastroenterology. 1997;113(6):1873–82.

    Article  CAS  PubMed  Google Scholar 

  89. Morampudi V, Conlin VS, Dalwadi U, Wu X, Marshall KC, Nguyen C, et al. Vasoactive intestinal peptide prevents PKCε-induced intestinal epithelial barrier disruption during EPEC infection. Am J Physiol Gastrointest Liver Physiol. 2015;308(5):G389–402.

    Article  CAS  PubMed  Google Scholar 

  90. Qin H, Zhang Z, Hang X, Jiang Y. L. plantarum prevents enteroinvasive Escherichia coli-induced tight junction proteins changes in intestinal epithelial cells. BMC Microbiol. 2009;9:63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Moriez R, Salvador-Cartier C, Theodorou V, Fioramonti J, Eutamene H, Bueno L. Myosin light chain kinase is involved in lipopolysaccharide-induced disruption of colonic epithelial barrier and bacterial translocation in rats. Am J Pathol. 2005;167(4):1071–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sheth P, Santos ND, Seth A, LaRusso NF, Rao RK. Lipopolysaccharide disrupts tight junctions in cholangiocyte monolayers by a c-Src-, TLR4-, and LBP-dependent mechanism. Am J Physiol Gastrointest Liver Physiol. 2007;293(1):G308–18.

    Article  CAS  PubMed  Google Scholar 

  93. Feng YH, Wang Y, Wang P, Huang YL, Wang FJ. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol Biochem. 2018;49(1):190–205.

    Article  CAS  PubMed  Google Scholar 

  94. Chen CL, Wang P, Su Q, Wang SL, Wang FJ. Myosin light chain kinase mediates intestinal barrier disruption following burn injury. PLoS One. 2012;7(4):e34946.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Samonte VA, Goto M, Ravindranath TM, Fazal N, Holloway VM, Goyal A, et al. Exacerbation of intestinal permeability in rats after a two-hit injury: burn and Enterococcus faecalis infection. Crit Care Med. 2004;32(11):2267–73.

    Article  PubMed  Google Scholar 

  96. Blikslager AT, Moeser AJ, Gookin JL, Jones SL, Odle J. Restoration of barrier function in injured intestinal mucosa. Physiol Rev. 2007;87(2):545–64.

    Article  CAS  PubMed  Google Scholar 

  97. Suzuki T. Regulation of intestinal epithelial permeability by tight junctions. Cell Mol Life Sci. 2013;70(4):631–59.

    Article  CAS  PubMed  Google Scholar 

  98. Turner JR, Rill BK, Carlson SL, Carnes D, Kerner R, Mrsny RJ, et al. Physiological regulation of epithelial tight junctions is associated with myosin light-chain phosphorylation. Am J Phys. 1997;273(4 Pt1):C1378–85.

    Article  CAS  Google Scholar 

  99. Somlyo AP, Somlyo AV. Signal transduction by G-proteins, Rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J Physiol. 2000;522:177–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Shen L, Black ED, Witkowski ED, Lencer WI, Guerriero V, Schneeberger EE, et al. Myosin light chain phosphorylation regulates barrier function by remodeling tight junction structure. J Cell Sci. 2006;119(10):2095–106.

    Article  CAS  PubMed  Google Scholar 

  101. Blair SA, Kane SV, Clayburgh DR, Turner JR. Epithelial myosin light chain kinase expression and activity are upregulated in inflammatory bowel disease. Lab Investig. 2006;86(2):191–201.

    Article  CAS  PubMed  Google Scholar 

  102. Turner JR. Molecular basis of epithelial barrier regulation: from basic mechanisms to clinical application. Am J Pathol. 2006;169(6):1901–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Wu CC, Lu YZ, Wu LL, Yu LC. Role of myosin light chain kinase in intestinal epithelial barrier defects in a rat model of bowel obstruction. BMC Gastroenterol. 2010;10:39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Gilbert S, Zhang R, Denson L, Moriggl R, Steinbrecher K, Shroyer N, et al. Enterocyte STAT5 promotes mucosal wound healing via suppression of myosin light chain kinase-mediated loss of barrier function and inflammation. EMBO Mol Med. 2012;4(1):1–16.

    Article  CAS  Google Scholar 

  105. Zahs A, Bird MD, Ramirez L, Turner JR, Choudhry MA, Kovacs EJ. Inhibition of long myosin light chain kinase activation alleviates intestinal damage after binge ethanol exposure and burn injury. Am J Physiol Gastrointest Liver Physiol. 2012;303(6):G705–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Guo M, Yuan SY, Sun C, Frederich BJ, Shen Q, McLean DL, et al. Role of non-muscle myosin light chain kinase in neutrophil-mediated intestinal barrier dysfunction during thermal injury. Shock. 2012;38(4):436–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Segain JP, Raingeard de la Bletiere D, Sauzeau V, Bourreille A, Hilaret G, Cario-Toumaniantz C, et al. Rho kinase blockade prevents inflammation via nuclear factor kappa B inhibition: evidence in Crohn’s disease and experimental colitis. Gastroenterology. 2003;124:1180–7.

    Article  CAS  PubMed  Google Scholar 

  108. Flynn AN, Buret AG. Tight junctional disruption and apoptosis in an in vitro model of Citrobacter rodentium infection. Microb Pathog. 2008;45(2):98–104.

    Article  CAS  PubMed  Google Scholar 

  109. Liu YL, Chen CL, Wang P, Wang FJ. Role of Rho kinase in regulating intestinal epithelial barrier dysfunction induced by sera from severely burned rata. J Med Postgrad. 2008;21(7):693–6.

    Google Scholar 

  110. Haines RJ, Wang CY, Yang CGY, Eitnier RA, Wang F, Wu MH. Targeting palmitoyl acyltransferase ZDHHC21 improves gut epithelial barrier dysfunction resulting from burn induced systemic inflammation. Am J Physiol Gastrointest Liver Physiol. 2017;313(6):G549–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Huang YL, Wang Y, Feng YH, Wang P, He XC, Ren H, Wang FJ. Role of endoplasmic reticulum stress-autophagy axis in severe burn-induced intestinal tight junction barrier dysfunction in mice. Front Physiol. 2019;10:606.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Cannon AR, Akhtar S, Hammer AM, Morris NL, Javorski MJ, Li X, et al. Effects of mesalamine treatment on gut barrier integrity after burn injury. J Burn Care Res. 2016;37(5):283–92.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Costantini TW, Peterson CY, Kroll L, Loomis WH, Eliceiri BP, Baird A, et al. Role of p38 MAPK in burn-induced intestinal barrier breakdown. J Surg Res. 2009;156(1):64–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Graham WV, He W, Marchiando AM, Zha J, Singh G, Li HS, Biswas A, et al. Intracellular MLCK1 diversion reverses barrier loss to restore mucosal homeostasis. Nat Med. 2019;25(4):690–700.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

This work was supported by the National Natural Science Foundation of China (81471871, 81772081).

Author information

Authors and Affiliations

Authors

Contributions

FW conceived and revised the review. WH and YW conducted the literature survey and drafted the review. PW participated in the literature survey. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Fengjun Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, W., Wang, Y., Wang, P. et al. Intestinal barrier dysfunction in severe burn injury. Burn Trauma 7, 24 (2019). https://doi.org/10.1186/s41038-019-0162-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41038-019-0162-3

Keywords