Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Dec 3.
Published in final edited form as: Annu Rev Immunol. 2012 Jan 3;30:149–173. doi: 10.1146/annurev-immunol-020711-075001

Microbial Translocation Across the GI Tract*

Jason M Brenchley 1, Daniel C Douek 2
PMCID: PMC3513328  NIHMSID: NIHMS420376  PMID: 22224779

Abstract

The lumen of the gastrointestinal (GI) tract is home to an enormous quantity of different bacterial species, our microbiota, that thrive in an often symbiotic relationship with the host. Given that the healthy host must regulate contact between the microbiota and its immune system to avoid overwhelming systemic immune activation, humans have evolved several mechanisms to attenuate systemic microbial translocation (MT) and its consequences. However, several diseases are associated with the failure of one or more of these mechanisms, with consequent immune activation and deleterious effects on health. Here, we discuss the mechanisms underlying MT, diseases associated with MT, and therapeutic interventions that aim to decrease it.

Keywords: intestinal permeability, innate immunity, inflammation

INTRODUCTION

The microbiota of the gastrointestinal (GI) tract comprises a large population of diverse bacterial species. The colon alone contains approximately 1014 microorganisms with approximately 1012 microorganisms per gram of colonic content. Thus, within an adult human the bacteria within the colon outnumber host cell numbers by up to two orders of magnitude, with a frequency of bacterial genes at least 100 times greater compared with those within the human genome. This microbiota is composed of approximately 1,000 species of predominantly unculturable bacteria that belong to two main phyla: the Firmicutes and the Bacteroidetes. Although the mechanistic details of the relationships between the microbiota and the host remain unclear, this relationship is undoubtedly complex and involves interactions among the individual members of the microbiota itself, the mucus layer of the GI tract, the local and systemic innate and adaptive immune systems, and the enterocytes.

Here, we discuss both beneficial and pathological interactions between the host and its microbiota, diseases that are characterized by unphysiological translocation of microbial products into peripheral circulation (microbial translocation, MT), mechanisms underlying MT, and therapeutic interventions that have been proposed to decrease pathologic MT.

INFLUENCES OF THE GI TRACT MICROBIOTA ON THE HOST

Local Relationships

Humans and the normal microbiota of the GI tract have evolved a symbiotic relationship. These beneficial interactions are highlighted by the finding that germ-free animals are more susceptible to infections and have reduced vascularity, digestive enzyme activity, muscle wall thickness, and serum immunoglobulin levels as well as smaller Peyer’s patches and fewer intraepithelial lymphocytes (reviewed in 1). Furthermore, these abnormalities appear to be enhanced in germ-free mice that were fed elemental diets containing no complex food antigens or bacterial products, and such animals also have lower numbers of circulating lymphocytes (2). The critical role of the microbiota is revealed by the finding that colonization of germ-free mice with a single species of bacterium is sufficient to enhance the mucosal immune system, including increased numbers of intraepithelial lymphocytes and increased activity of local antigen-presenting cells (APCs) (3). The beneficial roles that the GI microbiota play are multifactorial and may be separated into immunological, structural, and metabolic functions (4).

Being present in such high numbers, the organisms of the microbiota serve as competitors for potentially pathogenic bacterial species. This likely involves competition for limited sources of nutrition and limited sites of adherence to the epithelial barrier. The microbiota also protects against pathogenic infection by producing antimicrobial factors such as lactic acid, short chain fatty acids, and bacteriocins. Finally, the microbiota is capable of attenuating mucosal immune responses to pathogenic bacterial species. Indeed, certain microbiota species can promote nuclear export of the p65 segment of nuclear factor κB (NF-κB) through the peroxisome proliferator-activated receptor, thus limiting NF-κB-mediated transcription (5, 6).

The microbiota also leads to enhanced integrity of the structural barrier of the GI tract by metabolizing dietary carbohydrates into short-chain fatty acids, which are a major nutritional source for the colonic epithelia (7). Enterocytes also express Toll-like receptors (TLRs) through which signaling is thought to contribute to epithelial cell homeostasis (reviewed in 8). Consistent with this premise, decreased epithelial cell proliferation is observed in TLR-deficient mice (9). In addition to aiding in epithelial cell turnover, the microbiota is also involved in maintenance of the mucus layer. Indeed, the absence of the intestinal microbiota in germ-free mice is associated with a decrease in goblet cells, which are also smaller in size, and the thickness of the mucus layer is decreased (10).

In addition to the important role of the microbiota in maintaining the epithelial barrier and mucus layer, it also has a significant impact on production of luminal IgA. In the intestinal tract and other mucosal sites, most plasma cells secrete dimeric or oligomeric IgA that can transcytose directly into the lumen. That these antibodies are specific for luminal bacteria and viruses suggests that the microbiota shapes the specificity of luminal IgA (11). The corollary of this is exemplified in AID−/− mice, which do not produce sIgA and have a 100-fold increase in the number of small intestinal anaerobic bacteria (12). Secretory IgA thus serves as a line of defense against MT by limiting adhesion and entry into the epithelium, thereby facilitating clearance via the fecal stream (13, 14). Taken together, it is clear that the microbiota provides invaluable functions to the host locally within the GI tract.

Distal Relationships

In addition to the local effects that the microbiota has on the GI tract of the host, the microbiota also provides significant and beneficial functions for the host systems distal to the GI tract. Specifically, the microbiota metabolizes toxic and potentially carcinogenic compounds such as pyrolysates (15), thus reducing their bioavailability to the host. The microbiota also produces biotin, folate, and vitamin K from dietary precursors, which are then absorbed by the GI tract and circulated. That germ-free animals fed elemental diets have fewer systemic lymphocytes compared with conventional mice suggests a tonic stimulation of the systemic immune system by gut-derived microbial antigens (2).

Dysbiosis

Although the microbiota provides metabolites and beneficial immunological stimuli to the host, the actual composition of the microbiota also appears to have a significant impact on the host. Previous studies have implicated an altered balance in the composition of the microbiota (dysbiosis) in many diseases, such as obesity (16, 17), celiac disease (18), type 2 diabetes (19), atopic eczema (20, 21), asthma (22), inflammatory bowel disease (IBD) (23, 24), and chronic diarrhea (25).

Given the specific roles that the microbiota fulfills, it is reasonable to propose that its specific composition influences the capacity of the host to regulate its many functions. Indeed, only certain species of the microbiota, predominantly those belonging to a small subset of the Firmicutes, can metabolize complex carbohydrates into short-chain fatty acids (butyrate in particular) that can serve as growth factors for enterocytes (7). Along these lines, one study compared the composition of the microbiota in inflamed tissue and uninflamed tissue of individuals with IBD and tissue from healthy individuals and found fewer Firmicutes present in inflamed tissue from IBD patients (24). The same reduction in levels of Firmicutes was also observed within individuals diagnosed with type 2 diabetes, and the degree to which the microbiota was abnormal was correlated with plasma levels of glucose, suggesting that the dysbiotic microbiota may have a direct role in the pathogenesis of type 2 diabetes (19).

Additionally, in children diagnosed with atopic eczema and in non-breast-fed children, the microorganism Bifidobacterium pseudocatenulatum is more commonly identified than in healthy, breast-fed children, even though there are no correlations between severity of atopic eczema and levels of B. pseudocatenulatum (20).

Although low levels of Firmicutes can be associated with inflammatory conditions, their overgrowth is also associated with detrimental consequences. Increased levels of Firmicutes appear to alter the metabolic capacity of the microbiota, resulting in an increased ability to transfer carbohydrates, which results in host obesity (17).

In such correlative types of studies, it is difficult to conclude that an altered microbiota is causing disease rather than that the disease is affecting the composition of the microbiota. Indeed, among infants, factors such as geographical location, breast-feeding, mode of delivery, and antibiotic use can clearly alter the composition of the microbiota (26). Hence, alterations of the microbiota observed in disease states may be the result rather than the cause of disease. Comparative studies of culturable microbiota in human immunodeficiency virus (HIV)-infected and uninfected individuals have shown significant differences between the two, suggesting that the altered microbiota may contribute to HIV disease progression (27). Yet this finding could certainly be attributed to demographic differences between the two groups of individuals.

However, certain experimental approaches may distinguish between the two scenarios. For example, germ-free mice can be colonized with microbiota from diseased tissues or with microbiota of individuals suffering from diseases associated with altered microbiota. This approach has shown, for example, that microbiota from obese mice, transferred to germ-free animals, appears to cause the germ-free animals to gain significant weight (28). Alternatively, alterations in disease-associated microbiota through the use of probiotics and/or prebiotics could ameliorate symptoms of disease, as discussed in more detail below.

HOW THE MICROBIOTA IS EXCLUDED FROM SYSTEMIC CIRCULATION

The health of the host depends on the tight regulation of interactions between the host and microbiota. Translocation of microorganisms, or microorganism components, from the lumen of the GI tract into the systemic circulation can certainly have detrimental consequences, including activation of the immune system. In extreme cases of MT, septic shock ensues, where patient mortality can approach 70% (29) and is characterized by clinical manifestations including thermal dysregulation (hypothermia or hyperthermia), tachycardia, tachypnea, and altered white blood cell count (leukocytopenia or leukocytosis). Underlying these phenomena is an overwhelming production of inflammatory cytokines including tumor necrosis factor (TNF) and interleukin (IL)-1, and high motility group 1 protein (HMGB1) and nitric oxide. Although these trigger beneficial inflammatory responses to confine the infection and tissue damage, their excessive production results in elevated systemic inflammatory responses that may be more lethal than the bacterial infection itself (30). The importance of this phenomenon is of particular relevance in severe sepsis, where excessive production of proinflammatory mediators causes capillary leakage, tissue injury, and multiple organ failure (30). These proinflammatory mediators are predominantly produced by innate immune cells after stimulation through pattern-recognition receptors specific for bacterial products. Indeed, administration of bacterial lipopolysaccharide (LPS) in high doses is sufficient to recapitulate the physiologic abnormalities of septic shock (31). Thus, given the tremendous luminal bacterial burden, protecting against excessive MT may be regarded as essential to life.

Defense Against MT at the Gastrointestinal Surface

The first line of defense against MT is mediated by macromolecules within the lumen of the GI tract, including the constituents of the mucus layer: proteins, phospholipids, electrolytes, and water. The unique capacity of the mucus to protect the underlying epithelial surfaces is due primarily to the gel-forming properties of its glycoprotein mucins. Furthermore, luminal IgA and antimicrobial defensins can bind to and kill bacteria, thus limiting their ability to translocate.

Secondly, the epithelial barrier of the GI tract itself represents a significant obstacle against MT. There are four major types of GI tract epithelial cells (32): absorptive enterocytes; mucin-producing goblet cells; enteroendocrine cells, which produce peptide hormones (33); and Paneth cells, which secrete antimicrobial defensins, digestive enzymes, and growth factors (34). Enterocytes are short-lived cells, and the entire mouse GI epithelium is renewed every 3–4 days (35). Enterocytes are adjoined to one another via a complex of transmembrane and peripheral proteins that are tethered to the cytoskeleton of the adjacent enterocytes. These intercellular tight junctions are formed by interactions with claudin proteins, which form selective pores between enterocytes to promote specific ion permeability.

Should microbial products traverse the mucus and epithelial barriers, they are met by a large number of specialized resident macrophages that prevent such products from accessing the systemic circulation (36). The GI tract is a major reservoir of macrophages in the body (37) and have a very distinct phenotype and functional capacity. Although intestinal macrophages express high levels of HLA-DR and the myeloid marker aminopeptidase (CD13), similar to blood monocytes and other tissue macrophages (38), these cells are distinct in that they do not express the LPS coreceptor (CD14), the Fc receptors (CD89, CD16, CD32, and CD64), or receptors for IL-2 (CD25) and IL-3 (CD123) (39). The functional consequence of the absence of such receptors on intestinal macrophages is an inability to respond to many ligands that directly stimulate blood monocytes and other tissue macrophages. Indeed, GI macrophages do not produce proinflammatory cytokines such as IL-1 and TNF after stimulation with LPS (39). However, these cells can express various other pattern-recognition receptors including TLR4, TLR2, TLR5, and TLR9 and are capable of recognizing and phagocytosing bacterial antigens (38, 39). Hence, intestinal macrophages are specialized in their ability to clear antigens from the lamina propria without production of an inflammatory response to those antigens. This specialized role of intestinal macrophages is likely critically important to the maintenance of a noninflammatory state within the lamina propria of the GI tract.

Defense Against MT in the Liver

GI tract macrophages represent the first line of defense against translocated microbial products. However, should the GI tract macrophages fail to contain all such products, these are then drained by the portal vein into the liver. Thus, one of the many functions performed by the liver is the clearance of foreign and potentially harmful substances that drain from the GI tract. Indeed, concentrations of LPS in the portal vein are higher than in either the hepatic or peripheral veins (40), and bacteria can be cultured from healthy liver explants (41).

Besides the parenchymal hepatocytes, the liver contains other cell populations including liver sinusoidal endothelial cells (LSECs), tissue macrophages (Kupffer cells) and liver-associated lymphocytes. LSECs constitute the wall of the liver sinusoids, whereas Kupffer cells are located predominantly in the periportal area (42). Kupffer cells are therefore well situated for the phagocytosis of particulate antigens and organisms within the portal venous circulation. Both Kupffer cells and LSECs are responsive to direct stimulation with bacterial products (43), with measurably distinct responses compared with those of other tissue macrophages or monocytes in peripheral blood. Kupffer cells and LSECs constitutively express prostranoids and upregulate their expression concomitant with upregulation of IL-10 following LPS stimulation, which results in downregulation of antigen presentation by the APC within the liver (44, 45). Thus LPS-mediated stimulation of LSECs and Kupffer cells does not result in the release of proinflammatory mediators, and, similar to the response of GI tract macrophages, these cells are specialized in their ability to clear but not to respond immunologically to microbial antigens.

Defense Against MT in the Systemic Circulation

As discussed above, in order to access the systemic circulation, microbial antigens that originate in the GI tract must pass through the luminal mucus and IgA, traverse the tight epithelial barrier, escape uptake by GI tract lamina propria macrophages, and then avoid liver-mediated clearance by LSECs and Kupffer cells. Once in the circulation, microbial products are met with a further host-mediated response regulated by cell-surface receptors that sense and circulating factors that bind to and clear these products.

For example, healthy humans have high titers of circulating IgM, IgA, and IgG antibodies directed against the LPS core antigen that neutralize LPS activity (46, 47). When microbial products gain access to the circulation, such as during sepsis, these antibodies, termed EndoCAb, bind to and clear LPS from the circulation, and as a result their titers decrease (46). In contrast, in conditions when microbial products are found in the systemic circulation chronically, such as in IBD (discussed below), EndoCAb levels are increased (48), presumably as part of the normal humoral response to antigenic stimulation.

Additionally, the innate immune system produces soluble factors such as soluble CD14 (sCD14) and LPS-binding protein (LBP). CD14 is an LPS coreceptor expressed by peripheral blood monocytes and tissue macrophages. Following LPS stimulation, CD14+ monocytes/macrophages secrete sCD14 and shed surface CD14, which binds to LPS (49, 50). LBP, in contrast, is an acute phase reactant produced by hepatocytes (51). In healthy humans these proteins circulate at high concentrations in plasma, reaching the milligram/liter levels (50, 52). sCD14 and LBP both bind LPS and can, at given relative concentrations to one another and LPS, transfer LPS either to high-density lipoproteins (HDLs) to decrease the bioactivity of LPS or to the TLR4/MD-2/CD14 complex on monocytes/macrophages, leading to LPS-mediated stimulation (50). Indeed, the biological activities of different levels of sCD14 and LBP vis-à-vis LPS activity in vitro and in vivo are not completely understood, with experimental observations and interpretations varying considerably (50, 53). However, therapeutically increased levels of circulating LBP can be protective against gram-negative septicemia (54). Taken together, it is clear that several circulating factors, including EndoCAb, sCD14, LBP, and HDL, act as fundamental lines of defense against systemic stimulation of the immune system by translocated microbial antigens.

With approximately 1014 potential microorganisms residing within the GI tract at any given time, it is no surprise that humans allocate tremendous resources in corralling the microbes within the lumen of the GI tract and in controlling them should they enter the systemic circulation. Despite having these multiple mechanisms in place, the system could fail at any one of these checkpoints, and increased systemic MT would ensue.

MECHANISMS UNDERLYING MT

Low Levels of IgA

IgA is the most abundant antibody isotype in the body and is the second most dominant isotype in the peripheral circulation after IgG (55). IgA deficiency is the most common primary immunodeficiency (affecting between 1:300 and 1:3000 individuals) and has many genetic causes, including heavy chain gene deletions, T cell dysfunction, and alterations in cytokine signaling (56). Although many individuals with selective IgA deficiency are apparently asymptomatic, IgA-deficient individuals have a tendency to develop infections and disorders of the GI tract (57). Giardiasis, mal-absorption, lactose intolerance, celiac disease, ulcerative colitis (UC), nodular lymphoid hyperplasia, and increased epithelial cell proliferation are among the associated diseases (57, 58). Because the protective barrier of the GI system is impaired in IgA deficiency, protozoa such as Giardia lamblia can adhere to the epithelium, proliferate, and more easily cause infection (59). Even in the absence of infection, some GI tract luminal contents may enter the lamina propria and submucosal tissue. Indeed, individuals with IgA deficiency tend to mount large systemic IgG and IgM antibody responses to GI tract luminal antigens, including food and bacteria (60). Hence, low levels of IgA, with or without GI tract symptoms, can lead to increased MT; however, the degree to which MT occurs in IgA deficiency with subsequent systemic immune activation is unclear and warrants further study.

Alterations of the Structural Integrity of the GI Barrier

Pathogens

The tight epithelial barrier of the GI tract is clearly a major hurdle that must be breached in order for microbial antigens to traverse from the lumen of the intestine to the lamina propria of the GI tract. Consistent with this, many enterotoxins, which are expressed by pathogenic bacteria, target tight junction proteins of the GI tract. Enteropathogenic species of Vibrio, Escherichia, Salmonella, Helicobacter, and Clostridia all express such enterotoxins (6163). Moreover, it is thought that the ability of these bacteria to cause systemic infections is entirely dependent on enterotoxin expression. Thus, one could conclude that the disruption of tight junctions alone may cause increased MT. The importance of the tight epithelial barrier in protecting against MT is also highlighted by viral infections, which are associated with diarrheal diseases and subsequent increased intestinal permeability. Rotavirus, reovirus, norovirus, adenovirus, and coxsackievirus infections are all associated with disruption of the structural barrier of the GI tract and subsequent increased MT (64, 65).

Inflammation

In addition, alteration to the regulation of tight junction protein expression can lead to increased MT. Interferon (IFN)-γ expression increases claudin endocytosis with subsequent increase in paracellular permeability, whereas TNF and IL-13 lead to decreased expression of claudins (66, 67). Moreover, inflammatory conditions can be associated with upregulation of specific channel-forming claudins such as claudin 2 (66, 68). Indeed, it has been suggested that the ability of inflammatory cytokines to increase paracellular permeability allows neutrophil migration across epithelial barriers to combat invasive pathogens directly (69).

Not only can inflammatory cytokine production modulate claudin expression, but it can also influence the turnover of epithelial cells. Indeed, exposure to TNF or IL-1 can induce apoptosis in epithelial cells in vitro (70). Moreover, in vivo studies also suggest that excess production of TNF plays a deleterious role in perturbing the tight epithelial barrier, in part by induction of enterocyte apoptosis (reviewed in 71). Although inflammation may have multiple deleterious effects on GI tract integrity, it is important to note that therapeutic interventions aimed solely at decreasing TNF levels in vivo result in improved integrity of the structural barrier of the GI tract in individuals with CD (discussed below) (72).

TNF stimulation of enterocytes also leads to phosphorylation of the myosin light chain by myosin light chain kinase (MLCK) (73), which in turn leads to intestinal permeability via cytoskeleton rearrangement and modulation of tight junction protein expression. Consistent with this observation, inhibition of MLCK restores barrier function after TNF treatment and suggests that therapeutic interventions aimed at decreasing MLCK activity may be a promising approach in the treatment of TNF-mediated dysfunction of the structural barrier, as discussed below (74).

The propensity for chronic TNF signaling to induce GI tract structural damage via the mechanisms described above is highlighted by the finding that GI epithelial cells constitutively produce factors to extinguish TNF signaling. One such factor is A20, an NF-κB target gene that encodes a ubiquitin-editing enzyme essential for the termination of NF-κB activation after TNF or microbial product stimulation. Mice lacking A20 succumb to inflammation in several organs including the GI tract, and A20 mutations have been associated with CD (75). Moreover, tissue-specific disruption of A20 expression within GI tract enterocytes renders them exquisitely sensitive to TNF-induced toxicity and experimental colitis (75). Taken together, it is clear that inflammation may cause increased intestinal permeability and consequent MT.

Modulation of RORγt+ cells

Repair of the structural barrier of the GI tract after damage is also critically dependent on the local immune system. Certain lymphoid cells within the GI tract express the nuclear hormone receptor retinoic acid orphan receptor (ROR)γt and are involved in maintenance of the structural barrier of the GI tract and in defense against pathogens through the production of cytokines such as IL-17 and IL-22. IL-17 and IL-22 function in vivo to promote recruitment of neutrophils to areas of bacterial infection, to induce proliferation of enterocytes, and to produce defensins (7680). Although most data regarding IL-17-producing cells are derived from experiments in mice, several studies have shown that IL-17-producing cells can be identified in the blood of humans, can be characterized phenotypically based on expression patterns of certain chemokine and cytokine receptors, and appear to have specificity for bacterial and fungal antigens (8184). RORγt+ cells that produce IL-17 and IL-22 include CD4+ and CD8+ T cells and innate lymphoid cells (iLCs) such as lymphoid tissue–inducer cells and IL-22-producing NKp46+ cells (85).

Consistent with the notion that RORγt+ cells are important for maintenance of the structural barrier of the GI tract, mice lacking the transcription factor RORγt are significantly more prone to MT than wild-type mice (86). In these mice, containment of the luminal microbiota requires the generation of abnormally large numbers of tertiary lymphoid tissues (86). Although at steady state these animals tend to maintain the integrity of their GI tract, upon epithelial damage these mice have decreased regeneration of enterocytes and develop severe intestinal inflammation due to MT.

IL-17 production is also thought to play an important role in maintaining the appropriate immunological environment within the GI tract. A recent study described an accelerated wasting disease following induced colitis in mice incapable of IL-17 production (87). Moreover, in this model lack of IL-17 among responding T cells was sufficient to cause the effect. The mechanisms underlying this protective effect are likely multifaceted and include decreased neutrophil recruitment, decreased antibacterial defensin production, decreased regeneration of GI tract enterocytes, and increased frequencies of Th1-type CD4+ T cells, which produce high levels of tissue-damaging cytokines such as IFN-γ.

IL-22 is also thought to be critical for repair of the GI tract barrier. IL-22 belongs to the IL-10 family of cytokines (88), and its receptor is expressed on various epithelial tissues (89) and is believed to mediate epithelial innate immunity (90). The importance of IL-22 in repairing damage to the structural barrier of the GI tract is highlighted by studies of IL-22 knockout mice and by administration of IL-22-depleting antibodies. In these studies, mice that lack IL-22 are significantly more susceptible to chemically induced colitis (91).

IL-22 is clearly of lymphocyte origin with specific subsets of T cells (especially CD4 T cells) and iLCs capable of its production (92). However, the timing of IL-22 production may play an important role in the repair of the GI tract structural barrier, as is highlighted by experimental mouse infections with Citrobacter rodentium, which is characterized by damage to the structural barrier of the GI tract and MT. It was recently observed that rapid IL-22 production is critical for survival in this model, and it was suggested that IL-22 was produced by iLCs (93) in an IL-23-dependent manner. Therefore, though such studies suggest that IL-22 production by iLCs is critical for innate immunity in the intestine, the role of these cells in maintenance of the GI tract in the absence of infections is unclear given the relatively healthy structural barrier observed in IL-22-deficient mice (91) and the apparent overall GI tract health of uninfected mice selectively depleted of iLCs (93).

Decreased Microbial Clearance

In addition to mechanisms related to decreased ability to maintain the microbiota within the lumen of the GI tract leading to systemic MT, the inability to clear microbial products that cross the GI tract at steady state can also lead to systemic MT. The best studied mechanism related to decreased microbial product clearance is liver failure (discussed below). Consistent with this, there are several liver-associated diseases that are characterized by increased levels of microbial products in the peripheral circulation. Additionally, given that HDLs can clear LPS from the circulation, recent data suggest that the protective nature of high HDL levels against cardiovascular disease can be, at least partially, attributed to HDL’s ability to clear LPS from circulation, thus decreasing LPS-induced inflammation, which can accelerate atherogenesis (94). Consistent with a role for inflammation and decreased LPS clearance in cardiovascular disease, polymorphisms in the CD14 gene are associated with altered expression of sCD14 (95) and increased incidence of myocardial infarction (96). Although the levels of MT occurring at any given time in individuals without overt disease, but harboring mutations in CD14, LBP, and IgA or having low HDL levels, are currently unknown, the data clearly demonstrate that decreased clearance of microbial products from systemic circulation is associated with disease states.

DISEASES ASSOCIATED WITH SYSTEMIC MT

Given the numerous mechanisms that underlie the inability to restrict the microbiota to the GI lumen and the evidence suggesting that microbial products translocate frequently even at steady state, it comes as no surprise that there are multiple disease states that can be associated with translocation of microbial products into the peripheral circulation with consequent host responses.

Inflammatory Bowel Disease

MT has been most widely recognized as playing a major role in the pathogenesis of IBD. UC involves inflammation of the large bowel, whereas CD may involve inflammation of the entire GI tract. The etiology of IBD remains largely unknown, although mutations in genes encoding proteins involved in immunological responses through pattern-recognition receptors, genes involved in IL-17 production, and tight junction proteins are associated with IBD (97, 98). Additionally, altered composition of the GI tract microbiota has also been suggested to play a role in IBD pathogenesis (99, 100), as revealed by mouse models of IBD in which inflammation is significantly reduced when the mice are housed in germ-free conditions (101, 102).

Individuals with IBD also have elevated levels of circulating proinflammatory mediators (103105), and this systemic inflammation has been suggested to be due to MT (106108) because elevated serum levels of LPS (108113), bacterial DNA (114), EndoCAb (108, 112), and LBP (109, 112, 115) can be detected. In individuals with active disease, high levels of circulating bacterial products are associated with increased levels of proinflammatory cytokines (108, 112114), and granulocyte phagocytic activity is decreased, presumably due to recent bacterial phagocytosis in vivo (111). Finally, increased proinflammatory cytokine production by B cells has been suggested to be due to increased MT in individuals with IBD (113). That individuals with IBD have significantly higher intestinal permeability compared with healthy controls suggests that one of the mechanisms underlying disease pathogenesis in IBD is damage to the structural barrier of the GI tract (107). Taken together, it is clear that systemic MT occurs in IBD, and it follows that MT-induced immune activation may be at the heart of disease pathogenesis.

Human Immunodeficiency Virus Infection

During the acute phase of HIV infection, there is a significant insult to the immunological and structural components of the GI tract. Massive depletion of GI tract CD4 T cells (116120), low frequencies of IL-17-producing CD4 T cells (121123) and CD8 T cells (124), apoptosis of enterocytes (125), with subsequent damage to the structural barrier of the GI tract (126) and increased intestinal permeability (127130), are all manifestations of progressive HIV infection in humans and simian immunodeficiency virus (SIV) infection in Asian macaques. Moreover, generalized systemic activation of the immune system is a hallmark of the chronic phase of progressive HIV/SIV infection, and the degree to which the immune system is activated is the best predictor of the rate of disease progression (131135). Indeed, one cause of immune activation is increased MT due to damage to the GI tract, as elevated levels of LPS were found in the plasma of chronically HIV-infected individuals compared with either acutely HIV-infected or HIV-uninfected individuals (52, 136). Consistent with a proinflammatory role for LPS in the systemic circulation, levels of plasma LPS were associated with markers of immune activation of both the innate and adaptive arms of the immune system (52). Moreover, in patients with AIDS-associated dementia the activation status of monocytes in vivo was associated with levels of plasma LPS (137). Not only does MT occur during progressive HIV-1 infection, but increased levels of LPS are also observed in HIV-2-infected individuals (138).

Subsequently, using immunohistochemical analysis and the rhesus macaque/SIV model of HIV infection, investigators found that MT begins during the late acute phase of infection (~day 21–28 post infection), that microbial products colocalize with proinflammatory cytokines, and that one of the mechanisms underlying MT is damage to the structural barrier of the GI tract (126). The ability of the host to prevent microbial products from reaching circulation in the short term is highlighted by the normal plasma levels of LPS in acutely HIV-infected individuals, even though immunohistochemical analysis clearly demonstrates increased MT. Indeed, during the acute phase of infection EndoCAb titers decrease, sCD14 levels increase, and most microbial products found within the lamina propria of the GI tract are within the specialized tissue macrophages described above (52, 126, 139). In chronically infected individuals, GI tract macrophages fail to phagocytose all translocated bacterial products (126), EndoCAb titers remain low (52, 139, 140), and the number of Kupffer cells decreases (141). Hence, LPS clearance mechanisms are adversely affected during chronic HIV infection.

From these studies we can conclude that MT occurs in chronically HIV-infected individuals and that these microbial products can cause immune activation. It has also been proposed that the virus itself is a direct cause of immune activation in HIV infection (142, 143). However, there are certain cohorts of HIV-infected individuals in which viral replication is reduced to a minimum, yet immune activation remains pathologically elevated: elite controllers (ECs) and highly active antiretroviral therapy (HAART)–treated individuals. ECs are a rare group of individuals who spontaneously control viral replication to levels below the detection limit of conventional analyses (144, 145). Although these individuals have a significantly improved prognosis compared with viremic HIV-infected individuals, ECs tend to have higher levels of immune activation compared with HIV-uninfected individuals; many of these individuals nevertheless lose peripheral blood CD4 T cells, and some even progress to AIDS (133). In these individuals, elevated levels of LPS were detected and the frequency of activated phenotype CD38+ HLA-DR+ CD8 T cells correlated with MT (133). Moreover, there was a significant negative correlation between the levels of plasma LPS and the peripheral blood CD4 T cell count (133).

Additionally, recent studies have shown clearly that even though HAART can suppress plasma viral loads to undetectable levels, HAART-treated individuals nevertheless have increased mortality and morbidity compared with HIV-uninfected individuals, which are associated with inflammation and consequent cardiovascular disease (146, 147), osteopenia, (148), and cognitive decline (149). Given the long-term HAART-mediated control of viral replication in this group, it is unlikely that the residual inflammation is directly attributable to ongoing viral replication. Instead, a recent study suggests that elevated plasma levels of sCD14 independently predict increased mortality in HAART-treated, HIV-infected individuals (147). This is consistent with reports that, although chronic immune activation and levels of LPS in plasma decrease after initiating HAART, they remain elevated for years (52, 150152) and that GI tract CD4 T cells do not return to healthy levels even after long-term HAART (153155). In individuals with limited recovery of peripheral blood CD4 T cells after HAART, several studies have pointed to increased MT and immune activation as playing a causative role (52, 150152, 156158). Given recent data demonstrating increased mortality of HIV-infected individuals despite suppressed viral replication in HAART-treated individuals, and given the clear associations between mortality and inflammation and the associations with persistent MT, investigators have proposed (147) that adjunctive therapies aimed at reducing MT and/or its inflammatory consequences could improve the long-term prognosis of HIV-infected individuals.

Hepatitis B and C Virus Infection

Infection with hepatitis B (HBV) or C (HCV) virus can also be associated with increased systemic MT and immune activation. Infection with these hepatocytetropic viruses often leads to significant liver damage, increased systemic inflammation, and ultimately, liver fibrosis (159, 160). Consistent with decreased clearing of microbial products, Caradonna et al. (159) described increased plasma LPS levels in HCV-infected individuals that decreased after IFN-α treatment. That IFN-α treatment is associated with decreased MT and inflammation in chronically HCV-infected individuals suggests that IFN-α itself is unlikely to lead to damage to the structural barrier of the GI tract. Moreover, in patients with late-stage HCV-related cirrhosis, investigators found a concomitant increase in intestinal permeability and bacterial DNA within plasma (161). Finally, another study showed increased levels of plasma LPS, damage to the GI tract epithelial barrier, and increased sCD14 in patients with HBV or HCV infection (162). The levels of sCD14 in these patients correlated with markers of hepatic inflammation and fibrosis and predicted clinical outcome (162). The precise interplay between decreased microbial product clearing, systemic inflammation, intestinal permeability, and liver fibrosis in HCV-infected individuals is unclear, and further studies are certainly warranted.

Alcohol Use

Chronic alcohol use is also associated with significant inflammation and MT. There are two broad sources of alcohol-related inducers of inflammation: those derived from alcohol-damaged cells and those derived from the microbiota. Hypoxia, which results from alcohol metabolism, is known to induce an inflammatory response, but the underlying mechanisms remain unclear (163). However, MT has been extensively studied as a key inducer of inflammation in alcohol-related conditions. Alcoholics are known to have significantly elevated plasma LPS levels compared with healthy controls (164, 165). Indeed, heavy alcohol consumption is associated with an increase in gut permeability and MT independent of liver disease, and these effects are long lasting, with a two-week period of abstinence required for intestinal permeability to return to healthy levels (166). Moreover, acute heavy alcohol consumption is associated with a transient increase in plasma LPS in otherwise healthy human subjects (167), and animal models show that acute enteral alcohol administration to mice increases MT approximately fivefold within 30–90 min (168), whereas daily binge feeding of alcohol in rats for four weeks induces MT 15-fold compared with control animals (169).

The mechanisms underlying alcohol-induced MT are likely multifactorial. Recent studies have demonstrated that alcohol and/or acetaldehyde can directly increase gut permeability by induction of inducible nitric oxide synthase and NF-κB signaling, which, in turn, modulates a differential expression of tight junction proteins (170). Furthermore, damage to the liver could lead to decreased clearance of microbial products that translocate at steady state. Finally, chronic alcohol exposure also alters the composition of the microbiota, which results in bacterial overgrowth (171, 172). Consistent with MT playing a deleterious role in inflammation associated with chronic alcoholism, treatment with probiotics in alcoholic patients and animals with alcoholic liver disease resulted in decreased intestinal permeability and reduced liver tissue injury (173).

Fatty Liver Disease

A third liver-associated disease in which MT has been suggested to have a role is fatty liver disease (FLD) (174). Nonalcoholic FLD develops in the setting of obesity, insulin resistance, and high dietary carbohydrate intake (175). Although the specific etiology of FLD remains somewhat obscure, some have suggested that fatty livers are less capable of performing their normal functions (176). The decreased ability of the fatty liver to clear antigens and harmful substances from the circulation eventually leads to the death of hepatocytes, increased liver fibrosis, the accumulation of inflammatory cells within the liver, and systemic MT (177). Subsequently, MT then leads to increased inflammation and further liver damage, which perpetuates the cycle. Consistent with MT playing an important role in FLD, treatment of ob/ob mice (a model of FLD) with the nonorally absorbed antibiotic neomycin improves biological outcome (178).

Given the increased incidence of FLD in the setting of high dietary carbohydrate intake, it follows that the microbiota might be altered in individuals with FLD. Indeed, in a rat model of FLD, associated with total parenteral nutrition, the proliferation and overgrowth of certain gram-negative enteric organisms ensues (179). Hence, dysbiosis may play a role in MT, inflammation, decreased hepatic clearance, and increased liver fibrosis, which are associated with FLD. Consistent with MT and dysbiosis playing a role in FLD, treatment of a mouse model of FLD with probiotics leads to reduced hepatic fatty acid oxidation (180).

Pancreatitis

Acute pancreatitis has a mortality rate of approximately 10% with between 40% and 80% of the mortality due to sepsis (181, 182). Given that most bacteria associated with sepsis in pancreatitis are gram-negative enteric bacteria, it has been proposed that a series of events occurs in which, due to the proximity of the pancreas to the GI tract, local inflammation associated with acute pancreatitis results in damage to the structural barrier of the GI tract, increased intestinal permeability, and MT (182). Increased intestinal permeability as soon as 72 h after the onset of symptoms and the degree of intestinal permeability is directly associated with levels of LPS in the circulation (182). Moreover, increased MT has been observed in mouse models of acute pancreatitis (183). A subsequent study described increased intestinal permeability in individuals with severe compared with mild pancreatitis (184). In those individuals with severe pancreatitis, increased damage to the structural barrier of the GI tract is associated with increased plasma LPS and increased levels of circulating proinflammatory cytokines (184). Thus, many have suggested that therapeutic interventions for acute pancreatitis should also aim to decrease MT in affected individuals (181185).

Graft-versus-Host Disease

Given the rapid turnover of GI tract enterocytes, therapeutic interventions that aim to decrease proliferation of rapidly dividing cells result in damage to the structural barrier of the GI tract and systemic MT. Such is the case during the treatment of cancer with chemotherapeutic agents. Indeed, the MT that results from the conditioning regimen used for myeloablation before allogenic hematopoetic stem cell transplantation is thought to contribute to graft-versus-host disease (GVHD) (186). A role for LPS in the graft-versus-host response has been suggested by clinical studies aimed at decontamination of gram-negative bacteria from the GI tract during allogeneic stem cell transplantation, which has been shown to reduce GVHD (187), and the extent of such decontamination has been demonstrated to be an important predictor of GVHD severity (188). Notably, mutations in TLR4, which are associated with macrophage hyporesponsiveness to LPS within either the host or the donor, are associated with decreased incidence of GVHD (189). Similar results were seen in mouse models of GVHD when lymphocyte-depleted mice were reconstituted with allogenic stem cells from mice with mutations in TLR4 (190) or from CD14 knockout mice (191). Given the transient nature of damage to the GI tract after myeloablative chemotherapy, the role for MT in driving GVHD may be circumvented therapeutically, and antibiotics are one of several options. An alternative approach to limiting MT-induced immune activation is the administration of antibodies directed against microbial products. Indeed, infusion of a polyclonal antiserum against Escherichia coli as prophylaxis for acute GVHD in a prospective, placebo-controlled trial reduced overall GVHD from 63% to 42% and was found to be particularly efficacious in the subset of patients with severe GVHD (192).

THERAPEUTIC INTERVENTIONS TO DECREASE MT

As discussed, there are many levels at which both the host and microbiota minimize systemic immune activation from MT. Therefore, therapeutic interventions can be targeted against individual mechanisms underlying systemic MT. Such therapeutic interventions can be divided into four general classes: alteration of the composition of the microbiota, enhanced clearance of translocated microbial products, repair of the enterocyte barrier, and reduction of local inflammation.

Antibiotics

Possibly the most obvious therapeutic approach that might curb the deleterious effects of MT is nonabsorbed oral antibiotics. Indeed, use of a gut-sterilizing antibiotic regimen prior to abdominal surgery significantly decreases the incidence of subsequent wound infection and septicemia (193). The unique properties of rifaximin, a broad spectrum antibiotic that has low systemic absorption and high fecal concentrations, might, therefore, make it an ideal agent for the treatment of diseases associated with MT. Indeed, although rifaximin appears promising as a treatment for IBD, clinical trials to date have lacked sufficient power to assess its efficacy. In one multicenter, randomized, double-blind, placebo-controlled clinical trial, fewer treatment failures were seen in patients treated with rifaximin (n = 83). However, in a separate study there were no significant differences in clinical remission or improvement in active CD in patients receiving rifaximin compared with placebo (194).

Additionally, rifaximin may serve as a steroid-sparing agent for UC. In an open-label study of 30 patients receiving maintenance mesalamine in which rifaximin was used in lieu of steroids, approximately 77% of patients experienced clinical resolution (195). However, in another trial no significant clinical improvement with rifaximin compared with placebo was shown for patients with moderate-to-severe steroid-refractory UC (196). The disparate results from these clinical trials may have several explanations: nonoptimal rifaximin doses were used, the GI tract was unable to be sterilized for long periods of time, or there were differences in rifaximin-mediated alterations in the composition of the microbiota.

However, although decreasing the GI tract bacterial burden may improve the prognosis of individuals suffering from diseases associated with MT, as discussed above, the microbiota generally survives in a symbiotic relationship with the host. Although antibiotic use may decrease immune activation and improve some of the symptoms associated with MT-related diseases, long-term antibiotic use may not be the best therapeutic approach, as it results in outgrowth of antibiotic-resistant bacteria, decreased integrity of the structural barrier of the GI tract, and decreased bioavailability of microbiota-derived nutrients.

Probiotics

Given the ample data demonstrating that alterations to the composition of the microbiota often accompany diseases that are characterized by systemic MT and the described effects of probiotic organisms in maintaining the structural barrier of the GI tract, several studies have investigated the potential therapeutic benefit of promoting the growth of probiotic organisms. This goal has been pursued via direct oral administration of live probiotic organisms. Initially, studies were aimed at demonstrating proof of concept that orally administered live bacteria could survive transit through the length of the GI tract (197, 198). Although many bacterial species have been classified as probiotic, clinical trials showing any benefit to patients have been restricted to two mixtures of probiotic bacterial species: VSL#3 and Lactobacillus rhamnosus GG (207210). VSL#3—a combination of four strains of Lactobacilli, three strains of Bifidobacteria, and one strain of Streptococcus thermophillus—was shown to induce remission in 53% of treated individuals with UC (199). Moreover, a recent study demonstrated the safety and efficacy of VSL#3 in reducing symptoms of mild to moderate colitis with improved integrity of the structural barrier of the GI tract (200). The administration of Lactobacillus rhamnosus GG has shown clinical benefit in individuals with IBD (201, 202). Furthermore, a recent study described significantly increased reconstitution of peripheral blood CD4 T cells in chronically HIV-infected individuals treated with conventional antiretroviral therapy and L. rhamnosus (203). However, the potential effects of probiotics on improvement of the GI tract or on decreased immune activation were not studied. Finally, in animal models of FLD, probiotic supplementation reduced hepatic fatty acid oxidation (180). However, though large-scale placebo-controlled human trials are lacking, two small-scale human trials of probiotics for liver disease have been completed, and levels of liver enzymes were decreased among patients receiving probiotics (173, 204).

Antibodies Against Microbial Products

A second therapeutic approach to decreasing systemic MT is the administration of agents that clear microbial products from the circulation. Such compounds are generally monoclonal or polyclonal antibodies directed against microbial products. Historically, design of antimicrobial antigen immunoglobulin therapy was based upon preliminary data suggesting that mortality associated with sepsis was reduced by passive immunization with sera from individuals vaccinated with a mutant strain of Escherichia coli (205). Based on these findings, researchers developed several monoclonal antibodies directed against LPS for clinical trials of sepsis. The first were HA-1A and E5. These monoclonal antibodies and subsequently developed anti-LPS monoclonals have had only limited success in reducing mortality (reviewed in 206, 207). Because of the limited benefit afforded to septic individuals by administration of antibodies against microbial products, investigators have given little effort to studying such therapeutic approaches in settings of systemic MT. However, oral administration of a spray-dried, purified immunoglobulin protein isolate has been shown to decrease systemic inflammatory effects associated with MT in certain animal models (208210).

IL-22

Several therapeutic interventions aim to improve enterocyte homeostasis by the administration of cytokines such as IL-22, which is critical for maintenance of the structural barrier, particularly in the event of tissue damage (77, 85, 90, 211). Using a gene therapy approach, Sugimoto found that IL-22 administration could ameliorate intestinal inflammation in a mouse model of UC (212). This improvement was thought to be secondary to IL-22-enhanced mucus production and goblet cell replacement and restitution of the epithelial surface (212). Importantly, IL-22 receptor is also expressed by other epithelial cells, including those in the liver, and recombinant IL-22 administration resulted in decreased liver damage in a mouse model of hepatitis (213).

Glucagon-Like Peptide

Another epithelial cell growth factor which has been suggested as a therapeutic intervention to improve the structural integrity of the GI tract is glucagon-like peptide (GLP). GLP-2 is a 33 amino acid peptide produced with GLP-1 from the proglucagon gene, which encodes glucagon in the pancreas but undergoes specific posttranslational processing in the enteroendocrine L cells of the small intestine to produce the small GLP molecules. GLP-1 and -2 are released by L cells primarily in response to direct contact with luminal nutrients, especially long-chain fatty acids in the terminal ileum (214). Therapeutically, GLP-2 has been used extensively for treatment of short bowel syndrome with some success (reviewed in 215). Subsequently, certain GLP-2 analogs were developed and have been used to enhance the structural barrier of the GI tract in individuals with severe UC who were more likely to enter remission compared with placebo-treated individuals (216). Finally, GLP-2-treated individuals with CD had significantly improved enterocyte function compared with placebo-treated individuals (216).

CONCLUDING REMARKS

When considering the possible ramifications of harboring such an enormous bacterial burden within the GI tract, it seems reasonable to propose a few conclusions: (a) the interactions between the microbiota and host are generally symbiotic; (b) dysbiosis can both cause and result from systemic disease; (c) although humans have evolved multiple mechanisms to restrict the microbiota to the lumen of the GI tract, varying degrees of MT are a consistent feature in healthy humans; and (d ) chronic MT and consequent immune activation are a feature of many diseases. Indeed, multiple lines of evidence are consistent with each of these conclusions. Given the increasingly large number of studies that have demonstrated or proposed a role for MT in many pathologic processes in humans, it is clear that therapeutic interventions that mitigate MT and its effects on systemic immune activation could be of great clinical benefit to many individuals. However, it is also clear that multiple mechanisms can underlie an inability to contain microbial products completely within the lumen of the GI tract. Thus, the development of novel interventions that target MT will require a more detailed understanding of the molecular mechanisms that damage the integrity of the GI tract barrier, activate local immune responses, decrease clearance of translocated microbial products, activate systemic MT and immune responses, and perturb the composition of the microbiota.

Acknowledgments

We would like to acknowledge the members of the Cleveland Immunopathogenesis Consortium (BBC) for stimulating discussions.

Footnotes

*

This is a work of the U.S. Government and is not subject to copyright protection in the United States.

DISCLOSURE STATEMENT

The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

Contributor Information

Jason M. Brenchley, Email: jbrenchl@mail.nih.gov.

Daniel C. Douek, Email: ddouek@mail.nih.gov.

LITERATURE CITED

  • 1.Shanahan F. The host-microbe interface within the gut. Best Pract Res Clin Gastroenterol. 2002;16:915–31. doi: 10.1053/bega.2002.0342. [DOI] [PubMed] [Google Scholar]
  • 2.Wostmann BS, Pleasants JR, Bealmear P, Kincade PW. Serum proteins and lymphoid tissues in germ-free mice fed a chemically defined, water soluble, low molecular weight diet. Immunology. 1970;19:443–48. [PMC free article] [PubMed] [Google Scholar]
  • 3.Umesaki Y, Okada Y, Matsumoto S, Imaoka A, Setoyama H. Segmented filamentous bacteria are indigenous intestinal bacteria that activate intraepithelial lymphocytes and induce MHC class II molecules and fucosyl asialo GM1 glycolipids on the small intestinal epithelial cells in the ex-germ-free mouse. Microbiol Immunol. 1995;39:555–62. doi: 10.1111/j.1348-0421.1995.tb02242.x. [DOI] [PubMed] [Google Scholar]
  • 4.O’Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Rep. 2006;7:688–93. doi: 10.1038/sj.embor.7400731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Petrof EO, Kojima K, Ropeleski MJ, Musch MW, Tao Y, et al. Probiotics inhibit nuclear factor-κB and induce heat shock proteins in colonic epithelial cells through proteasome inhibition. Gastroenterology. 2004;127:1474–87. doi: 10.1053/j.gastro.2004.09.001. [DOI] [PubMed] [Google Scholar]
  • 6.Kelly D, Campbell JI, King TP, Grant G, Jansson EA, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-γ and RelA. Nat Immunol. 2004;5:104–12. doi: 10.1038/ni1018. [DOI] [PubMed] [Google Scholar]
  • 7.Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA: acetate CoA-transferase gene. Environ Microbiol. 2010;12:304–14. doi: 10.1111/j.1462-2920.2009.02066.x. [DOI] [PubMed] [Google Scholar]
  • 8.Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol. 2010;10:131–44. doi: 10.1038/nri2707. [DOI] [PubMed] [Google Scholar]
  • 9.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–41. doi: 10.1016/j.cell.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 10.Kandori H, Hirayama K, Takeda M, Doi K. Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice. Exp Anim. 1996;45:155–60. doi: 10.1538/expanim.45.155. [DOI] [PubMed] [Google Scholar]
  • 11.Brandtzaeg P, Baekkevold ES, Farstad IN, Jahnsen FL, Johansen FE, et al. Regional specialization in the mucosal immune system: What happens in the microcompartments? Immunol Today. 1999;20:141–51. doi: 10.1016/s0167-5699(98)01413-3. [DOI] [PubMed] [Google Scholar]
  • 12.Fagarasan S, Muramatsu M, Suzuki K, Nagaoka H, Hiai H, Honjo T. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science. 2002;298:1424–27. doi: 10.1126/science.1077336. [DOI] [PubMed] [Google Scholar]
  • 13.Johansen FE, Pekna M, Norderhaug IN, Haneberg B, Hietala MA, et al. Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J Exp Med. 1999;190:915–22. doi: 10.1084/jem.190.7.915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science. 2004;303:1662–65. doi: 10.1126/science.1091334. [DOI] [PubMed] [Google Scholar]
  • 15.Morotomi M, Mutai M. In vitro binding of potent mutagenic pyrolysates to intestinal bacteria. J Natl Cancer Inst. 1986;77:195–201. [PubMed] [Google Scholar]
  • 16.Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–84. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
  • 18.De Palma G, Nadal I, Medina M, Donat E, Ribes-Koninckx C, et al. Intestinal dysbiosis and reduced immunoglobulin-coated bacteria associated with coeliac disease in children. BMC Microbiol. 2010;10:63. doi: 10.1186/1471-2180-10-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE. 2010;5:e9085. doi: 10.1371/journal.pone.0009085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gore C, Munro K, Lay C, Bibiloni R, Morris J, et al. Bifidobacterium pseudocatenulatum is associated with atopic eczema: a nested case-control study investigating the fecal microbiota of infants. J Allergy Clin Immunol. 2008;121:135–40. doi: 10.1016/j.jaci.2007.07.061. [DOI] [PubMed] [Google Scholar]
  • 21.Penders J, Stobberingh EE, van den Brandt PA, Thijs C. The role of the intestinal microbiota in the development of atopic disorders. Allergy. 2007;62:1223–36. doi: 10.1111/j.1398-9995.2007.01462.x. [DOI] [PubMed] [Google Scholar]
  • 22.Huffnagle GB. The microbiota and allergies/asthma. PLoS Pathog. 2010;6:e1000549. doi: 10.1371/journal.ppat.1000549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tannock GW. Molecular analysis of the intestinal microflora in IBD. Mucosal Immunol. 2008;1(Suppl 1):S15–18. doi: 10.1038/mi.2008.54. [DOI] [PubMed] [Google Scholar]
  • 24.Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2010;11:7. doi: 10.1186/1471-2180-11-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Swidsinski A, Loening-Baucke V, Verstraelen H, Osowska S, Doerffel Y. Biostructure of fecal microbiota in healthy subjects and patients with chronic idiopathic diarrhea. Gastroenterology. 2008;135:568–79. doi: 10.1053/j.gastro.2008.04.017. [DOI] [PubMed] [Google Scholar]
  • 26.Fallani M, Young D, Scott J, Norin E, Amarri S, et al. Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics. J Pediatr Gastroenterol Nutr. 2010;51:77–84. doi: 10.1097/MPG.0b013e3181d1b11e. [DOI] [PubMed] [Google Scholar]
  • 27.Gori A, Tincati C, Rizzardini G, Torti C, Quirino T, et al. Early impairment of gut function and gut flora supporting a role for alteration of gastrointestinal mucosa in human immunodeficiency virus pathogenesis. J Clin Microbiol. 2008;46:757–58. doi: 10.1128/JCM.01729-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–23. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Matot I, Sprung CL. Definition of sepsis. Intensive Care Med. 2001;27(Suppl 1):S3–9. doi: 10.1007/pl00003795. [DOI] [PubMed] [Google Scholar]
  • 30.Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348:138–50. doi: 10.1056/NEJMra021333. [DOI] [PubMed] [Google Scholar]
  • 31.Lindsey DC, Emerson TE, Jr, Thompson TE, John AE, Duerr ML, et al. Characterization of an endotoxemic baboon model of metabolic and organ dysfunction. Circ Shock. 1991;34:298–310. [PubMed] [Google Scholar]
  • 32.Loeffler M, Birke A, Winton D, Potten C. Somatic mutation, monoclonality and stochastic models of stem cell organization in the intestinal crypt. J Theor Biol. 1993;160:471–91. doi: 10.1006/jtbi.1993.1031. [DOI] [PubMed] [Google Scholar]
  • 33.Roth KA, Hertz JM, Gordon JI. Mapping enteroendocrine cell populations in transgenic mice reveals an unexpected degree of complexity in cellular differentiation within the gastrointestinal tract. J Cell Biol. 1990;110:1791–801. doi: 10.1083/jcb.110.5.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bry L, Falk P, Huttner K, Ouellette A, Midtvedt T, Gordon JI. Paneth cell differentiation in the developing intestine of normal and transgenic mice. Proc Natl Acad Sci USA. 1994;91:10335–39. doi: 10.1073/pnas.91.22.10335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gordon JI, Hermiston ML. Differentiation and self-renewal in the mouse gastrointestinal epithelium. Curr Opin Cell Biol. 1994;6:795–803. doi: 10.1016/0955-0674(94)90047-7. [DOI] [PubMed] [Google Scholar]
  • 36.Smith PD, Ochsenbauer-Jambor C, Smythies LE. Intestinal macrophages: unique effector cells of the innate immune system. Immunol Rev. 2005;206:149–59. doi: 10.1111/j.0105-2896.2005.00288.x. [DOI] [PubMed] [Google Scholar]
  • 37.Lee SH, Starkey PM, Gordon S. Quantitative analysis of total macrophage content in adult mouse tissues. Immunochemical studies with monoclonal antibody F4/80. J Exp Med. 1985;161:475–89. doi: 10.1084/jem.161.3.475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Smith PD, Smythies LE, Mosteller-Barnum M, Sibley DA, Russell MW, et al. Intestinal macrophages lack CD14 and CD89 and consequently are down-regulated for LPS- and IgA-mediated activities. J Immunol. 2001;167:2651–56. doi: 10.4049/jimmunol.167.5.2651. [DOI] [PubMed] [Google Scholar]
  • 39.Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Investig. 2005;115:66–75. doi: 10.1172/JCI19229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lumsden AB, Henderson JM, Kutner MH. Endotoxin levels measured by a chromogenic assay in portal, hepatic and peripheral venous blood in patients with cirrhosis. Hepatology. 1988;8:232–36. doi: 10.1002/hep.1840080207. [DOI] [PubMed] [Google Scholar]
  • 41.Singh R, Bullard J, Kalra M, Assefa S, Kaul AK, et al. Status of bacterial colonization, Toll-like receptor expression and nuclear factor-κB activation in normal and diseased human livers. Clin Immunol. 2011;138:41–49. doi: 10.1016/j.clim.2010.09.006. [DOI] [PubMed] [Google Scholar]
  • 42.Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS. The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology. 1985;5:683–92. doi: 10.1002/hep.1840050427. [DOI] [PubMed] [Google Scholar]
  • 43.Shnyra A, Lindberg AA. Scavenger receptor pathway for lipopolysaccharide binding to Kupffer and endothelial liver cells in vitro. Infect Immun. 1995;63:865–73. doi: 10.1128/iai.63.3.865-873.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Grewe M, Duyster J, Dieter P, Henninger H, Schulze-Specking A, Decker K. Prostaglandin D2 and E2 syntheses in rat Kupffer cells are antagonistically regulated by lipopolysaccharide and phorbol ester. Biol Chem Hoppe Seyler. 1992;373:655–64. doi: 10.1515/bchm3.1992.373.2.655. [DOI] [PubMed] [Google Scholar]
  • 45.Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Buschenfelde KH, Gerken G. Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol. 1995;22:226–92. doi: 10.1016/0168-8278(95)80433-1. [DOI] [PubMed] [Google Scholar]
  • 46.Strutz F, Heller G, Krasemann K, Krone B, Muller GA. Relationship of antibodies to endotoxin core to mortality in medical patients with sepsis syndrome. Intensive Care Med. 1999;25:435–44. doi: 10.1007/s001340050877. [DOI] [PubMed] [Google Scholar]
  • 47.Cohen IR, Norins LC. Natural human antibodies to gram-negative bacteria: immunoglobulins G, A, and M. Science. 1966;152:1257–59. doi: 10.1126/science.152.3726.1257. [DOI] [PubMed] [Google Scholar]
  • 48.Barclay GR. Endogenous endotoxin-core antibody (EndoCAb) as a marker of endotoxin exposure and a prognostic indicator: a review. Prog Clin Biol Res. 1995;392:263–72. [PubMed] [Google Scholar]
  • 49.Bazil V, Strominger JL. Shedding as a mechanism of down-modulation of CD14 on stimulated human monocytes. J Immunol. 1991;147:1567–74. [PubMed] [Google Scholar]
  • 50.Kitchens RL, Thompson PA. Modulatory effects of sCD14 and LBP on LPS-host cell interactions. J Endotoxin Res. 2005;11:225–29. doi: 10.1179/096805105X46565. [DOI] [PubMed] [Google Scholar]
  • 51.Ulevitch RJ, Tobias PS. Receptor-dependent mechanisms of cell stimulation by bacterial endotoxin. Annu Rev Immunol. 1995;13:437–57. doi: 10.1146/annurev.iy.13.040195.002253. [DOI] [PubMed] [Google Scholar]
  • 52.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–71. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 53.Tapping RI, Gegner JA, Kravchenko VV, Tobias PS. Roles for LBP and soluble CD14 in cellular uptake of LPS. Prog Clin Biol Res. 1998;397:73–78. [PubMed] [Google Scholar]
  • 54.Lamping N, Dettmer R, Schroder NW, Pfeil D, Hallatschek W, et al. LPS-binding protein protects mice from septic shock caused by LPS or gram-negative bacteria. J Clin Investig. 1998;101:2065–71. doi: 10.1172/JCI2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kerr MA. The structure and function of human IgA. Biochem J. 1990;271:285–96. doi: 10.1042/bj2710285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yel L. Selective IgA deficiency. J Clin Immunol. 2010;30:10–16. doi: 10.1007/s10875-009-9357-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Edwards E, Razvi S, Cunningham-Rundles C. IgA deficiency: clinical correlates and responses to pneumococcal vaccine. Clin Immunol. 2004;111:93–97. doi: 10.1016/j.clim.2003.12.005. [DOI] [PubMed] [Google Scholar]
  • 58.Meini A, Pillan NM, Villanacci V, Monafo V, Ugazio AG, Plebani A. Prevalence and diagnosis of celiac disease in IgA-deficient children. Ann Allergy Asthma Immunol. 1996;77:333–36. doi: 10.1016/S1081-1206(10)63329-7. [DOI] [PubMed] [Google Scholar]
  • 59.Zinneman HH, Kaplan AP. The association of giardiasis with reduced intestinal secretory immunoglobulin A. Am J Dig Dis. 1972;17:793–97. doi: 10.1007/BF02231148. [DOI] [PubMed] [Google Scholar]
  • 60.Cardinale F, Friman V, Carlsson B, Bjorkander J, Armenio L, Hanson LA. Aberrations in titre and avidity of serum IgM and IgG antibodies to microbial and food antigens in IgA deficiency. Scand J Immunol. 1992;36:279–83. doi: 10.1111/j.1365-3083.1992.tb03100.x. [DOI] [PubMed] [Google Scholar]
  • 61.Wu Z, Nybom P, Magnusson KE. Distinct effects of Vibrio cholerae haemagglutinin/protease on the structure and localization of the tight junction-associated proteins occludin and ZO-1. Cell Microbiol. 2000;2:11–17. doi: 10.1046/j.1462-5822.2000.00025.x. [DOI] [PubMed] [Google Scholar]
  • 62.Fedwick JP, Lapointe TK, Meddings JB, Sherman PM, Buret AG. Helicobacter pylori activates myosin light-chain kinase to disrupt claudin-4 and claudin-5 and increase epithelial permeability. Infect Immun. 2005;73:7844–52. doi: 10.1128/IAI.73.12.7844-7852.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hopkins AM, Walsh SV, Verkade P, Boquet P, Nusrat A. Constitutive activation of Rho proteins by CNF-1 influences tight junction structure and epithelial barrier function. J Cell Sci. 2003;116:725–42. doi: 10.1242/jcs.00300. [DOI] [PubMed] [Google Scholar]
  • 64.Guttman JA, Finlay BB. Tight junctions as targets of infectious agents. Biochim Biophys Acta. 2009;1788:832–41. doi: 10.1016/j.bbamem.2008.10.028. [DOI] [PubMed] [Google Scholar]
  • 65.Troeger H, Loddenkemper C, Schneider T, Schreier E, Epple HJ, et al. Structural and functional changes of the duodenum in human norovirus infection. Gut. 2009;58:1070–77. doi: 10.1136/gut.2008.160150. [DOI] [PubMed] [Google Scholar]
  • 66.Prasad S, Mingrino R, Kaukinen K, Hayes KL, Powell RM, et al. Inflammatory processes have differential effects on claudins 2, 3 and 4 in colonic epithelial cells. Lab Investig. 2005;85:1139–62. doi: 10.1038/labinvest.3700316. [DOI] [PubMed] [Google Scholar]
  • 67.Chiba H, Kojima T, Osanai M, Sawada N. The significance of interferon-γ-triggered internalization of tight-junction proteins in inflammatory bowel disease. Sci STKE. 2006;2006(316):pe1. doi: 10.1126/stke.3162006pe1. [DOI] [PubMed] [Google Scholar]
  • 68.Smith AJ, Schacker TW, Reilly CS, Haase AT. A role for syndecan-1 and claudin-2 in microbial translocation during HIV-1 infection. J Acquir Immune Defic Syndr. 2010;55:306–15. doi: 10.1097/QAI.0b013e3181ecfeca. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Walsh SV, Hopkins AM, Nusrat A. Modulation of tight junction structure and function by cytokines. Adv Drug Deliv Rev. 2000;41:303–13. doi: 10.1016/s0169-409x(00)00048-x. [DOI] [PubMed] [Google Scholar]
  • 70.Ruemmele FM, Beaulieu JF, Dionne S, Levy E, Seidman EG, et al. Lipopolysaccharide modulation of normal enterocyte turnover by Toll-like receptors is mediated by endogenously produced tumour necrosis factor α. Gut. 2002;51:842–48. doi: 10.1136/gut.51.6.842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Grunfeld C, Palladino MA., Jr Tumor necrosis factor: immunologic, antitumor, metabolic, and cardiovascular activities. Adv Intern Med. 1990;35:45–71. [PubMed] [Google Scholar]
  • 72.Tilg H, Kaser A. Antitumour necrosis factor therapy in Crohn’s disease. Expert Opin Biol Ther. 2002;2:715–21. doi: 10.1517/14712598.2.7.715. [DOI] [PubMed] [Google Scholar]
  • 73.Clayburgh DR, Barrett TA, Tang Y, Meddings JB, Van Eldik LJ, et al. Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo. J Clin Investig. 2005;115:2702–15. doi: 10.1172/JCI24970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zolotarevsky Y, Hecht G, Koutsouris A, Gonzalez DE, Quan C, et al. A membrane-permeant peptide that inhibits MLC kinase restores barrier function in in vitro models of intestinal disease. Gastroenterology. 2002;123:163–72. doi: 10.1053/gast.2002.34235. [DOI] [PubMed] [Google Scholar]
  • 75.Vereecke L, Sze M, Mc Guire C, Rogiers B, Chu Y, et al. Enterocyte-specific A20 deficiency sensitizes to tumor necrosis factor-induced toxicity and experimental colitis. J Exp Med. 2010;207:1513–23. doi: 10.1084/jem.20092474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kao CY, Chen Y, Thai P, Wachi S, Huang F, et al. IL-17 markedly up-regulates beta-defensin-2 expression in human airway epithelium via JAK and NF-κB signaling pathways. J Immunol. 2004;173:3482–91. doi: 10.4049/jimmunol.173.5.3482. [DOI] [PubMed] [Google Scholar]
  • 77.Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203:2271–79. doi: 10.1084/jem.20061308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kolls JK, Linden A. Interleukin-17 family members and inflammation. Immunity. 2004;21:467–76. doi: 10.1016/j.immuni.2004.08.018. [DOI] [PubMed] [Google Scholar]
  • 79.Huang W, Na L, Fidel PL, Schwarzenberger P. Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis. 2004;190:624–31. doi: 10.1086/422329. [DOI] [PubMed] [Google Scholar]
  • 80.Chung DR, Kasper DL, Panzo RJ, Chitnis T, Grusby MJ, et al. CD4+ T cells mediate abscess formation in intra-abdominal sepsis by an IL-17-dependent mechanism. J Immunol. 2003;170:1958–63. doi: 10.4049/jimmunol.170.4.1958. [DOI] [PubMed] [Google Scholar]
  • 81.Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007;8:639–46. doi: 10.1038/ni1467. [DOI] [PubMed] [Google Scholar]
  • 82.Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204:1849–61. doi: 10.1084/jem.20070663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sato W, Aranami T, Yamamura T. Cutting edge: human Th17 cells are identified as bearing CCR2+CCR5- phenotype. J Immunol. 2007;178:7525–29. doi: 10.4049/jimmunol.178.12.7525. [DOI] [PubMed] [Google Scholar]
  • 84.Milner JD, Brenchley JM, Laurence A, Freeman AF, Hill BJ, et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 2008;452:773–76. doi: 10.1038/nature06764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sawa S, Lochner M, Satoh-Takayama N, Dulauroy S, Berard M, et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol. 2011;12:320–26. doi: 10.1038/ni.2002. [DOI] [PubMed] [Google Scholar]
  • 86.Lochner M, Ohnmacht C, Presley L, Bruhns P, Si-Tahar M, et al. Microbiota-induced tertiary lymphoid tissues aggravate inflammatory disease in the absence of RORγt and LTi cells. J Exp Med. 2010;208:125–34. doi: 10.1084/jem.20100052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.O’Connor W, Jr, Kamanaka M, Booth CJ, Town T, Nakae S, et al. A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol. 2009;10:603–9. doi: 10.1038/ni.1736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 2004;22:929–79. doi: 10.1146/annurev.immunol.22.012703.104622. [DOI] [PubMed] [Google Scholar]
  • 89.Gurney AL. IL-22, a Th1 cytokine that targets the pancreas and select other peripheral tissues. Int Immunopharmacol. 2004;4:669–77. doi: 10.1016/j.intimp.2004.01.016. [DOI] [PubMed] [Google Scholar]
  • 90.Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. IL-22 increases the innate immunity of tissues. Immunity. 2004;21:241–54. doi: 10.1016/j.immuni.2004.07.007. [DOI] [PubMed] [Google Scholar]
  • 91.Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med. 2009;206:1465–72. doi: 10.1084/jem.20082683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009;206:35–41. doi: 10.1084/jem.20072713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sonnenberg GF, Monticelli LA, Elloso MM, Fouser LA, Artis D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity. 2011;34:122–34. doi: 10.1016/j.immuni.2010.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Suzuki M, Pritchard DK, Becker L, Hoofnagle AN, Tanimura N, et al. High-density lipoprotein suppresses the type I interferon response, a family of potent antiviral immunoregulators, in macrophages challenged with lipopolysaccharide. Circulation. 2010;122:1919–27. doi: 10.1161/CIRCULATIONAHA.110.961193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Poikonen K, Lajunen T, Silvennoinen-Kassinen S, Paldanius M, Leinonen M, Saikku P. Susceptibility of human monocyte-macrophages to Chlamydia pneumoniae infection in vitro is highly variable and associated with levels of soluble CD14 and C. pneumoniae IgA and human HSP-IgG antibodies in serum. Scand J Immunol. 2008;67:279–84. doi: 10.1111/j.1365-3083.2007.02061.x. [DOI] [PubMed] [Google Scholar]
  • 96.Hubacek JA, Rothe G, Pit’ha J, Skodova Z, Stanek V, et al. C(−260)–>T polymorphism in the promoter of the CD14 monocyte receptor gene as a risk factor for myocardial infarction. Circulation. 1999;99:3218–20. doi: 10.1161/01.cir.99.25.3218. [DOI] [PubMed] [Google Scholar]
  • 97.Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;347:417–29. doi: 10.1056/NEJMra020831. [DOI] [PubMed] [Google Scholar]
  • 98.Sartor RB. Pathogenesis and immune mechanisms of chronic inflammatory bowel diseases. Am J Gastroenterol. 1997;92:5S–11S. [PubMed] [Google Scholar]
  • 99.Elson CO, Cong Y, McCracken VJ, Dimmitt RA, Lorenz RG, Weaver CT. Experimental models of inflammatory bowel disease reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev. 2005;206:260–76. doi: 10.1111/j.0105-2896.2005.00291.x. [DOI] [PubMed] [Google Scholar]
  • 100.Obermeier F, Dunger N, Strauch UG, Hofmann C, Bleich A, et al. CpG motifs of bacterial DNA essentially contribute to the perpetuation of chronic intestinal inflammation. Gastroenterology. 2005;129:913–27. doi: 10.1053/j.gastro.2005.06.061. [DOI] [PubMed] [Google Scholar]
  • 101.Harper PH, Lee EC, Kettlewell MG, Bennett MK, Jewell DP. Role of the faecal stream in the maintenance of Crohn’s colitis. Gut. 1985;26:279–84. doi: 10.1136/gut.26.3.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224–31. doi: 10.1128/iai.66.11.5224-5231.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Targan SR. Biology of inflammation in Crohn’s disease: mechanisms of action of anti-TNF-α therapy. Can J Gastroenterol. 2000;14(Suppl C):13C–16C. doi: 10.1155/2000/409396. [DOI] [PubMed] [Google Scholar]
  • 104.Schreiber S, Nikolaus S, Hampe J, Hamling J, Koop I, et al. Tumour necrosis factor α and interleukin 1β in relapse of Crohn’s disease. Lancet. 1999;353:459–61. doi: 10.1016/S0140-6736(98)03339-X. [DOI] [PubMed] [Google Scholar]
  • 105.Vermeire S, Van Assche G, Rutgeerts P. C-reactive protein as a marker for inflammatory bowel disease. Inflamm Bowel Dis. 2004;10:661–65. doi: 10.1097/00054725-200409000-00026. [DOI] [PubMed] [Google Scholar]
  • 106.Papp M, Foldi I, Altorjay I, Palyu E, Udvardy M, et al. Anti-microbial antibodies in celiac disease: trick or treat? World J Gastroenterol. 2009;15:3891–900. doi: 10.3748/wjg.15.3891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hollander D, Vadheim CM, Brettholz E, Petersen GM, Delahunty T, Rotter JI. Increased intestinal permeability in patients with Crohn’s disease and their relatives. A possible etiologic factor. Ann Intern Med. 1986;105:883–85. doi: 10.7326/0003-4819-105-6-883. [DOI] [PubMed] [Google Scholar]
  • 108.Gardiner KR, Halliday MI, Barclay GR, Milne L, Brown D, et al. Significance of systemic endotoxaemia in inflammatory bowel disease. Gut. 1995;36:897–901. doi: 10.1136/gut.36.6.897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Pastor Rojo O, Lopez San Roman A, Albeniz Arbizu E, de la Hera Martinez A, Ripoll Sevillano E, Albillos Martinez A. Serum lipopolysaccharide-binding protein in endotoxemic patients with inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:269–77. doi: 10.1002/ibd.20019. [DOI] [PubMed] [Google Scholar]
  • 110.Caradonna L, Amati L, Magrone T, Pellegrino NM, Jirillo E, Caccavo D. Enteric bacteria, lipopolysaccharides and related cytokines in inflammatory bowel disease: biological and clinical significance. J Endotoxin Res. 2000;6:205–14. [PubMed] [Google Scholar]
  • 111.Caradonna L, Amati L, Lella P, Jirillo E, Caccavo D. Phagocytosis, killing, lymphocyte-mediated antibacterial activity, serum autoantibodies, and plasma endotoxins in inflammatory bowel disease. Am J Gastroenterol. 2000;95:1495–502. doi: 10.1111/j.1572-0241.2000.02085.x. [DOI] [PubMed] [Google Scholar]
  • 112.Pasternak BA, D’Mello S, Jurickova II, Han X, Willson T, et al. Lipopolysaccharide exposure is linked to activation of the acute phase response and growth failure in pediatric Crohn’s disease and murine colitis. Inflamm Bowel Dis. 2010;16:856–69. doi: 10.1002/ibd.21132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.McDonnell M, Liang Y, Noronha A, Coukos J, Kasper DL, et al. Systemic Toll-like receptor ligands modify B-cell responses in human inflammatory bowel disease. Inflamm Bowel Dis. 2010;17:298–307. doi: 10.1002/ibd.21424. [DOI] [PubMed] [Google Scholar]
  • 114.Gutierrez A, Frances R, Amoros A, Zapater P, Garmendia M, et al. Cytokine association with bacterial DNA in serum of patients with inflammatory bowel disease. Inflamm Bowel Dis. 2009;15:508–14. doi: 10.1002/ibd.20806. [DOI] [PubMed] [Google Scholar]
  • 115.Lakatos PL, Kiss LS, Palatka K, Altorjay I, Antal-Szalmas P, et al. Serum lipopolysaccharide-binding protein and soluble CD14 are markers of disease activity in patients with Crohn’s disease. Inflamm Bowel Dis. 2011;17:767–77. doi: 10.1002/ibd.21402. [DOI] [PubMed] [Google Scholar]
  • 116.Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature. 2005;434:1093–97. doi: 10.1038/nature03501. [DOI] [PubMed] [Google Scholar]
  • 117.Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200:749–59. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, et al. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science. 1998;280:427–31. doi: 10.1126/science.280.5362.427. [DOI] [PubMed] [Google Scholar]
  • 119.Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–70. doi: 10.1084/jem.20041196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol. 2003;77:11708–17. doi: 10.1128/JVI.77.21.11708-11717.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, et al. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood. 2008;112:2826–35. doi: 10.1182/blood-2008-05-159301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Favre D, Lederer S, Kanwar B, Ma ZM, Proll S, et al. Critical loss of the balance between Th17 and T regulatory cell populations in pathogenic SIV infection. PLoS Pathog. 2009;5:e1000295. doi: 10.1371/journal.ppat.1000295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cecchinato V, Trindade C, Laurence A, Heraud J, Brenchley J, et al. Altered balance between Th17 and Th1 cells at mucosal sites predicts AIDS progression in simian immunodeficiency virus-infected macaques. Mucosal Immunol. 2008;1:279–88. doi: 10.1038/mi.2008.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Nigam P, Kwa S, Velu V, Amara RR. Loss of IL-17-producing CD8 T cells during late chronic stage of pathogenic simian immunodeficiency virus infection. J Immunol. 2010;186:745–53. doi: 10.4049/jimmunol.1002807. [DOI] [PubMed] [Google Scholar]
  • 125.Li Q, Estes JD, Duan L, Jessurun J, Pambuccian S, et al. Simian immunodeficiency virus-induced intestinal cell apoptosis is the underlying mechanism of the regenerative enteropathy of early infection. J Infect Dis. 2008;197:420–29. doi: 10.1086/525046. [DOI] [PubMed] [Google Scholar]
  • 126.Estes JD, Harris LD, Klatt NR, Tabb B, Pittaluga S, et al. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLoS Pathog. 2010;6:e1001052. doi: 10.1371/journal.ppat.1001052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Sharpstone D, Neild P, Crane R, Taylor C, Hodgson C, et al. Small intestinal transit, absorption, and permeability in patients with AIDS with and without diarrhoea. Gut. 1999;45:70–76. doi: 10.1136/gut.45.1.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kapembwa MS, Fleming SC, Sewankambo N, Serwadda D, Lucas S, et al. Altered small-intestinal permeability associated with diarrhoea in human-immunodeficiency-virus-infected Caucasian and African subjects. Clin Sci. 1991;81:327–34. doi: 10.1042/cs0810327. [DOI] [PubMed] [Google Scholar]
  • 129.Clayton F, Kapetanovic S, Kotler DP. Enteric microtubule depolymerization in HIV infection: a possible cause of HIV-associated enteropathy. AIDS. 2001;15:123–24. doi: 10.1097/00002030-200101050-00019. [DOI] [PubMed] [Google Scholar]
  • 130.Kotler DP, Gaetz HP, Lange M, Klein EB, Holt PR. Enteropathy associated with the acquired immunodeficiency syndrome. Ann Intern Med. 1984;101:421–28. doi: 10.7326/0003-4819-101-4-421. [DOI] [PubMed] [Google Scholar]
  • 131.Giorgi JV, Hultin LE, McKeating JA, Johnson TD, Owens B, et al. Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis. 1999;179:859–70. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
  • 132.Giorgi JV, Lyles RH, Matud JL, Yamashita TE, Mellors JW, et al. Predictive value of immunologic and virologic markers after long or short duration of HIV-1 infection. J Acquir Immune Defic Syndr. 2002;29:346–55. doi: 10.1097/00126334-200204010-00004. [DOI] [PubMed] [Google Scholar]
  • 133.Hunt PW, Brenchley JM, Sinclair E, McCune JM, Roland M, et al. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis. 2008;197:126–33. doi: 10.1086/524143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187:1534–43. doi: 10.1086/374786. [DOI] [PubMed] [Google Scholar]
  • 135.Deeks SG, Kitchen CM, Liu L, Guo H, Gascon R, et al. Immune activation set point during early HIV infection predicts subsequent CD4+ T-cell changes independent of viral load. Blood. 2004;104:942–47. doi: 10.1182/blood-2003-09-3333. [DOI] [PubMed] [Google Scholar]
  • 136.Brenchley JM, Price DA, Douek DC. HIV disease: fallout from a mucosal catastrophe? Nat Immunol. 2006;7:235–39. doi: 10.1038/ni1316. [DOI] [PubMed] [Google Scholar]
  • 137.Ancuta P, Kamat A, Kunstman KJ, Kim EY, Autissier P, et al. Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients. PLoS ONE. 2008;3:e 2516. doi: 10.1371/journal.pone.0002516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Nowroozalizadeh S, Mansson F, da Silva Z, Repits J, Dabo B, et al. Microbial translocation correlates with the severity of both HIV-1 and HIV-2 infections. J Infect Dis. 2010;201:1150–54. doi: 10.1086/651430. [DOI] [PubMed] [Google Scholar]
  • 139.Papasavvas E, Pistilli M, Reynolds G, Bucki R, Azzoni L, et al. Delayed loss of control of plasma lipopolysaccharide levels after therapy interruption in chronically HIV-1-infected patients. AIDS. 2009;23:369–75. doi: 10.1097/QAD.0b013e32831e9c76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Balagopal A, Philp FH, Astemborski J, Block TM, Mehta A, et al. Human immunodeficiency virus-related microbial translocation and progression of hepatitis C. Gastroenterology. 2008;135:226–33. doi: 10.1053/j.gastro.2008.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Balagopal A, Ray SC, De Oca RM, Sutcliffe CG, Vivekanandan P, et al. Kupffer cells are depleted with HIV immunodeficiency and partially recovered with antiretroviral immune reconstitution. AIDS. 2009;23:2397–404. doi: 10.1097/QAD.0b013e3283324344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Beignon AS, McKenna K, Skoberne M, Manches O, DaSilva I, et al. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via Toll-like receptor-viral RNA interactions. J Clin Investig. 2005;115:3265–75. doi: 10.1172/JCI26032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Meier A, Chang JJ, Chan ES, Pollard RB, Sidhu HK, et al. Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat Med. 2009;15:955–59. doi: 10.1038/nm.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Vesanen M, Stevens CE, Taylor PE, Rubinstein P, Saksela K. Stability in controlling viral replication identifies long-term nonprogressors as a distinct subgroup among human immunodeficiency virus type 1-infected persons. J Virol. 1996;70:9035–40. doi: 10.1128/jvi.70.12.9035-9040.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Migueles SA, Sabbaghian MS, Shupert WL, Bettinotti MP, Marincola FM, et al. HLA B*5701 is highly associated with restriction of virus replication in a subgroup of HIV-infected long term nonprogressors. Proc Natl Acad Sci USA. 2000;97:2709–14. doi: 10.1073/pnas.050567397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, et al. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 2008;5:e203. doi: 10.1371/journal.pmed.0050203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Sandler NG, Wand H, Roque A, Law M, Nason MC, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203:780–90. doi: 10.1093/infdis/jiq118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Arnsten JH, Freeman R, Howard AA, Floris-Moore M, Lo Y, Klein RS. Decreased bone mineral density and increased fracture risk in aging men with or at risk for HIV infection. AIDS. 2007;21:617–23. doi: 10.1097/QAD.0b013e3280148c05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.McCutchan JA, Wu JW, Robertson K, Koletar SL, Ellis RJ, et al. HIV suppression by HAART preserves cognitive function in advanced, immune-reconstituted AIDS patients. AIDS. 2007;21:1109–17. doi: 10.1097/QAD.0b013e3280ef6acd. [DOI] [PubMed] [Google Scholar]
  • 150.Anselmi A, Vendrame D, Rampon O, Giaquinto C, Zanchetta M, De Rossi A. Immune reconstitution in human immunodeficiency virus type 1-infected children with different virological responses to anti-retroviral therapy. Clin Exp Immunol. 2007;150:442–50. doi: 10.1111/j.1365-2249.2007.03526.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Massanella M, Negredo E, Perez-Alvarez N, Puig J, Ruiz-Hernandez R, et al. CD4 T-cell hyperactivation and susceptibility to cell death determine poor CD4 T-cell recovery during suppressive HAART. AIDS. 2010;24:959–68. doi: 10.1097/QAD.0b013e328337b957. [DOI] [PubMed] [Google Scholar]
  • 152.Baroncelli S, Galluzzo CM, Pirillo MF, Mancini MG, Weimer LE, et al. Microbial translocation is associated with residual viral replication in HAART-treated HIV+ subjects with <50copies/ml HIV-1 RNA. J Clin Virol. 2009;46:367–70. doi: 10.1016/j.jcv.2009.09.011. [DOI] [PubMed] [Google Scholar]
  • 153.Mehandru S, Poles MA, Tenner-Racz K, Jean-Pierre P, Manuelli V, et al. Lack of mucosal immune reconstitution during prolonged treatment of acute and early HIV-1 infection. PLoS Med. 2006;3:e484. doi: 10.1371/journal.pmed.0030484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Guadalupe M, Sankaran S, George MD, Reay E, Verhoeven D, et al. Viral suppression and immune restoration in the gastrointestinal mucosa of human immunodeficiency virus type 1-infected patients initiating therapy during primary or chronic infection. J Virol. 2006;80:8236–47. doi: 10.1128/JVI.00120-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Macal M, Sankaran S, Chun TW, Reay E, Flamm J, et al. Effective CD4+ T-cell restoration in gut-associated lymphoid tissue of HIV-infected patients is associated with enhanced Th17 cells and polyfunctional HIV-specific T-cell responses. Mucosal Immunol. 2008;1:475–88. doi: 10.1038/mi.2008.35. [DOI] [PubMed] [Google Scholar]
  • 156.Cassol E, Malfeld S, Mahasha P, van der Merwe S, Cassol S, et al. Persistent microbial translocation and immune activation in HIV-1-infected South Africans receiving combination antiretroviral therapy. J Infect Dis. 2010;202:723–33. doi: 10.1086/655229. [DOI] [PubMed] [Google Scholar]
  • 157.Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, et al. Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis. 2009;199:1177–85. doi: 10.1086/597476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Marchetti G, Bellistri GM, Borghi E, Tincati C, Ferramosca S, et al. Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS. 2008;22:2035–38. doi: 10.1097/QAD.0b013e3283112d29. [DOI] [PubMed] [Google Scholar]
  • 159.Caradonna L, Mastronardi ML, Magrone T, Cozzolongo R, Cuppone R, et al. Biological and clinical significance of endotoxemia in the course of hepatitis C virus infection. Curr Pharm Des. 2002;8:995–1005. doi: 10.2174/1381612024606983. [DOI] [PubMed] [Google Scholar]
  • 160.Neuman MG, Sha K, Esguerra R, Zakhari S, Winkler RE, et al. Inflammation and repair in viral hepatitis C. Dig Dis Sci. 2008;53:1468–87. doi: 10.1007/s10620-007-0047-3. [DOI] [PubMed] [Google Scholar]
  • 161.Thalheimer U, De Iorio F, Capra F, del Mar Lleo M, Zuliani V, et al. Altered intestinal function precedes the appearance of bacterial DNA in serum and ascites in patients with cirrhosis: a pilot study. Eur J Gastroenterol Hepatol. 2010;22:1228–34. doi: 10.1097/MEG.0b013e32833b4b03. [DOI] [PubMed] [Google Scholar]
  • 162.Sandler NG, Koh C, Roque A, Eccleston JL, Siegel RB, et al. Host response to translocated microbial products predicts outcomes of patients with HBV or HCV infection. Gastroenterology. 2011;141:1220–30. doi: 10.1053/j.gastro.2011.06.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Oliver KM, Taylor CT, Cummins EP. Hypoxia. Regulation of NFκB signalling during inflammation: the role of hydroxylases. Arthritis Res Ther. 2009;11:215. doi: 10.1186/ar2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Fukui H, Brauner B, Bode JC, Bode C. Plasma endotoxin concentrations in patients with alcoholic and non-alcoholic liver disease: reevaluation with an improved chromogenic assay. J Hepatol. 1991;12:162–69. doi: 10.1016/0168-8278(91)90933-3. [DOI] [PubMed] [Google Scholar]
  • 165.Schafer C, Parlesak A, Schutt C, Bode JC, Bode C. Concentrations of lipopolysaccharide-binding protein, bactericidal/permeability-increasing protein, soluble CD14 and plasma lipids in relation to endotoxaemia in patients with alcoholic liver disease. Alcohol Alcohol. 2002;37:81–86. doi: 10.1093/alcalc/37.1.81. [DOI] [PubMed] [Google Scholar]
  • 166.Bjarnason I, Peters TJ, Wise RJ. The leaky gut of alcoholism: possible route of entry for toxic compounds. Lancet. 1984;1:179–82. doi: 10.1016/s0140-6736(84)92109-3. [DOI] [PubMed] [Google Scholar]
  • 167.Bode C, Kugler V, Bode JC. Endotoxemia in patients with alcoholic and non-alcoholic cirrhosis and in subjects with no evidence of chronic liver disease following acute alcohol excess. J Hepatol. 1987;4:8–14. doi: 10.1016/s0168-8278(87)80003-x. [DOI] [PubMed] [Google Scholar]
  • 168.Rivera CA, Bradford BU, Seabra V, Thurman RG. Role of endotoxin in the hypermetabolic state after acute ethanol exposure. Am J Physiol. 1998;275:G1252–58. doi: 10.1152/ajpgi.1998.275.6.G1252. [DOI] [PubMed] [Google Scholar]
  • 169.Enomoto N, Yamashina S, Kono H, Schemmer P, Rivera CA, et al. Development of a new, simple rat model of early alcohol-induced liver injury based on sensitization of Kupffer cells. Hepatology. 1999;29:1680–89. doi: 10.1002/hep.510290633. [DOI] [PubMed] [Google Scholar]
  • 170.Bode C, Bode JC. Effect of alcohol consumption on the gut. Best Pract Res Clin Gastroenterol. 2003;17:575–92. doi: 10.1016/s1521-6918(03)00034-9. [DOI] [PubMed] [Google Scholar]
  • 171.Bode JC, Bode C, Heidelbach R, Durr HK, Martini GA. Jejunal microflora in patients with chronic alcohol abuse. Hepatogastroenterology. 1984;31:30–34. [PubMed] [Google Scholar]
  • 172.Hauge T, Persson J, Danielsson D. Mucosal bacterial growth in the upper gastrointestinal tract in alcoholics (heavy drinkers) Digestion. 1997;58:591–95. doi: 10.1159/000201507. [DOI] [PubMed] [Google Scholar]
  • 173.Kirpich IA, Solovieva NV, Leikhter SN, Shidakova NA, Lebedeva OV, et al. Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: a pilot study. Alcohol. 2008;42:675–82. doi: 10.1016/j.alcohol.2008.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Abu-Shanab A, Quigley EM. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2010;7:691–701. doi: 10.1038/nrgastro.2010.172. [DOI] [PubMed] [Google Scholar]
  • 175.Solga S, Alkhuraishe AR, Clark JM, Torbenson M, Greenwald A, et al. Dietary composition and nonalcoholic fatty liver disease. Dig Dis Sci. 2004;49:1578–83. doi: 10.1023/b:ddas.0000043367.69470.b7. [DOI] [PubMed] [Google Scholar]
  • 176.Yang SQ, Lin HZ, Lane MD, Clemens M, Diehl AM. Obesity increases sensitivity to endotoxin liver injury: implications for the pathogenesis of steatohepatitis. Proc Natl Acad Sci USA. 1997;94:2557–62. doi: 10.1073/pnas.94.6.2557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Ruiz AG, Casafont F, Crespo J, Cayon A, Mayorga M, et al. Lipopolysaccharide-binding protein plasma levels and liver TNF-α gene expression in obese patients: evidence for the potential role of endotoxin in the pathogenesis of non-alcoholic steatohepatitis. Obes Surg. 2007;17:1374–80. doi: 10.1007/s11695-007-9243-7. [DOI] [PubMed] [Google Scholar]
  • 178.Koteish A, Diehl AM. Animal models of steatosis. Semin Liver Dis. 2001;21:89–104. doi: 10.1055/s-2001-12932. [DOI] [PubMed] [Google Scholar]
  • 179.Pappo I, Becovier H, Berry EM, Freund HR. Polymyxin B reduces cecal flora, TNF production and hepatic steatosis during total parenteral nutrition in the rat. J Surg Res. 1991;51:106–12. doi: 10.1016/0022-4804(91)90078-z. [DOI] [PubMed] [Google Scholar]
  • 180.Li Z, Yang S, Lin H, Huang J, Watkins PA, et al. Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology. 2003;37:343–50. doi: 10.1053/jhep.2003.50048. [DOI] [PubMed] [Google Scholar]
  • 181.Beger HG, Bittner R, Block S, Buchler M. Bacterial contamination of pancreatic necrosis. A prospective clinical study. Gastroenterology. 1986;91:433–38. doi: 10.1016/0016-5085(86)90579-2. [DOI] [PubMed] [Google Scholar]
  • 182.Ammori BJ, Leeder PC, King RF, Barclay GR, Martin IG, et al. Early increase in intestinal permeability in patients with severe acute pancreatitis: correlation with endotoxemia, organ failure, and mortality. J Gastrointest Surg. 1999;3:252–62. doi: 10.1016/s1091-255x(99)80067-5. [DOI] [PubMed] [Google Scholar]
  • 183.Vasilescu C, Herlea V, Buttenschoen K, Beger HG. Endotoxin translocation in two models of experimental acute pancreatitis. J Cell Mol Med. 2003;7:417–24. doi: 10.1111/j.1582-4934.2003.tb00244.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Penalva JC, Martinez J, Laveda R, Esteban A, Munoz C, et al. A study of intestinal permeability in relation to the inflammatory response and plasma Endocab IgM levels in patients with acute pancreatitis. J Clin Gastroenterol. 2004;38:512–17. doi: 10.1097/01.mcg.0000129060.46654.e0. [DOI] [PubMed] [Google Scholar]
  • 185.Ammori BJ, Fitzgerald P, Hawkey P, McMahon MJ. The early increase in intestinal permeability and systemic endotoxin exposure in patients with severe acute pancreatitis is not associated with systemic bacterial translocation: molecular investigation of microbial DNA in the blood. Pancreas. 2003;26:18–22. doi: 10.1097/00006676-200301000-00004. [DOI] [PubMed] [Google Scholar]
  • 186.Langrehr JM, Machens C, Zill E, Leder K, Nussler A, et al. Bacterial translocation during graft-versus-host disease after small bowel transplantation is reduced following inhibition of inducible nitric oxide synthesis. Transplantation. 2000;69:2415–21. doi: 10.1097/00007890-200006150-00035. [DOI] [PubMed] [Google Scholar]
  • 187.Storb R, Prentice RL, Buckner CD, Clift RA, Appelbaum F, et al. Graft-versus-host disease and survival in patients with aplastic anemia treated by marrow grafts from HLA-identical siblings. Beneficial effect of a protective environment. N Engl J Med. 1983;308:302–7. doi: 10.1056/NEJM198302103080602. [DOI] [PubMed] [Google Scholar]
  • 188.Beelen DW, Haralambie E, Brandt H, Linzenmeier G, Muller KD, et al. Evidence that sustained growth suppression of intestinal anaerobic bacteria reduces the risk of acute graft-versus-host disease after sibling marrow transplantation. Blood. 1992;80:2668–76. [PubMed] [Google Scholar]
  • 189.Lorenz E, Schwartz DA, Martin PJ, Gooley T, Lin MT, et al. Association of TLR4 mutations and the risk for acute GVHD after HLA-matched-sibling hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2001;7:384–87. doi: 10.1053/bbmt.2001.v7.pm11529488. [DOI] [PubMed] [Google Scholar]
  • 190.Cooke KR, Hill GR, Crawford JM, Bungard D, Brinson YS, et al. Tumor necrosis factor-α production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft-versus-host disease. J Clin Investig. 1998;102:1882–91. doi: 10.1172/JCI4285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Cooke KR, Olkiewicz K, Erickson N, Ferrara JL. The role of endotoxin and the innate immune response in the pathophysiology of acute graft versus host disease. J Endotoxin Res. 2002;8:441–48. doi: 10.1179/096805102125001046. [DOI] [PubMed] [Google Scholar]
  • 192.Bayston K, Baumgartner JD, Clark P, Cohen J. Anti-endotoxin antibody for prevention of acute GVHD. Bone Marrow Transplant. 1991;8:426–27. [PubMed] [Google Scholar]
  • 193.Alexander-Williams J, McLeish AR, Keighley KR, Burdon DW. Prophylactic antibiotics in bowel surgery. Proc R Soc Med. 1976;69:327–29. [PMC free article] [PubMed] [Google Scholar]
  • 194.Prantera C, Lochs H, Campieri M, Scribano ML, Sturniolo GC, et al. Antibiotic treatment of Crohn’s disease: results of a multicentre, double blind, randomized, placebo-controlled trial with rifaximin. Aliment Pharmacol Ther. 2006;23:1117–25. doi: 10.1111/j.1365-2036.2006.02879.x. [DOI] [PubMed] [Google Scholar]
  • 195.Guslandi M, Petrone MC, Testoni PA. Rifaximin for active ulcerative colitis. Inflamm Bowel Dis. 2006;12:335. doi: 10.1097/01.MIB.0000215092.85116.6c. [DOI] [PubMed] [Google Scholar]
  • 196.Gionchetti P, Rizzello F, Ferrieri A, Venturi A, Brignola C, et al. Rifaximin in patients with moderate or severe ulcerative colitis refractory to steroid-treatment: a double-blind, placebo-controlled trial. Dig Dis Sci. 1999;44:1220–21. doi: 10.1023/a:1026648812439. [DOI] [PubMed] [Google Scholar]
  • 197.Reid G, Charbonneau D, Erb J, Kochanowski B, Beuerman D, et al. Oral use of Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 significantly alters vaginal flora: randomized, placebo-controlled trial in 64 healthy women. FEMS Immunol Med Microbiol. 2003;35:131–34. doi: 10.1016/S0928-8244(02)00465-0. [DOI] [PubMed] [Google Scholar]
  • 198.Morelli L, Zonenenschain D, Del Piano M, Cognein P. Utilization of the intestinal tract as a delivery system for urogenital probiotics. J Clin Gastroenterol. 2004;38:S107–10. doi: 10.1097/01.mcg.0000128938.32835.98. [DOI] [PubMed] [Google Scholar]
  • 199.Bibiloni R, Fedorak RN, Tannock GW, Madsen KL, Gionchetti P, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100:1539–46. doi: 10.1111/j.1572-0241.2005.41794.x. [DOI] [PubMed] [Google Scholar]
  • 200.Sood A, Midha V, Makharia GK, Ahuja V, Singal D, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7:1202–9. 9, e1. doi: 10.1016/j.cgh.2009.07.016. [DOI] [PubMed] [Google Scholar]
  • 201.Zocco MA, dal Verme LZ, Cremonini F, Piscaglia AC, Nista EC, et al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther. 2006;23:1567–74. doi: 10.1111/j.1365-2036.2006.02927.x. [DOI] [PubMed] [Google Scholar]
  • 202.Gupta P, Andrew H, Kirschner BS, Guandalini S. Is Lactobacillus GG helpful in children with Crohn’s disease? Results of a preliminary, open-label study. J Pediatr Gastroenterol Nutr. 2000;31:453–57. doi: 10.1097/00005176-200010000-00024. [DOI] [PubMed] [Google Scholar]
  • 203.Irvine SL, Hummelen R, Hekmat S, Looman CW, Habbema JD, Reid G. Probiotic yogurt consumption is associated with an increase of CD4 count among people living with HIV/AIDS. J Clin Gastroenterol. 2010;44:e201–5. doi: 10.1097/MCG.0b013e3181d8fba8. [DOI] [PubMed] [Google Scholar]
  • 204.Loguercio C, De Simone T, Federico A, Terracciano F, Tuccillo C, et al. Gut-liver axis: a new point of attack to treat chronic liver damage? Am J Gastroenterol. 2002;97:2144–64. doi: 10.1111/j.1572-0241.2002.05942.x. [DOI] [PubMed] [Google Scholar]
  • 205.Ziegler EJ, McCutchan JA, Fierer J, Glauser MP, Sadoff JC, et al. Treatment of gram-negative bacteremia and shock with human antiserum to a mutant Escherichia coli. N Engl J Med. 1982;307:1225–30. doi: 10.1056/NEJM198211113072001. [DOI] [PubMed] [Google Scholar]
  • 206.Baumgartner JD. Monoclonal anti-endotoxin antibodies for the treatment of gram-negative bacteremia and septic shock. Eur J Clin Microbiol Infect Dis. 1990;9:711–16. doi: 10.1007/BF02184682. [DOI] [PubMed] [Google Scholar]
  • 207.Barclay GR. Endotoxin-core antibodies: time for a reappraisal? Intensive Care Med. 1999;25:427–29. doi: 10.1007/s001340050874. [DOI] [PubMed] [Google Scholar]
  • 208.Perez-Bosque A, Miro L, Polo J, Russell L, Campbell J, et al. Dietary plasma protein supplements prevent the release of mucosal proinflammatory mediators in intestinal inflammation in rats. J Nutr. 2010;140:25–30. doi: 10.3945/jn.109.112466. [DOI] [PubMed] [Google Scholar]
  • 209.Corl BA, Harrell RJ, Moon HK, Phillips O, Weaver EM, et al. Effect of animal plasma proteins on intestinal damage and recovery of neonatal pigs infected with rotavirus. J Nutr Biochem. 2007;18:778–84. doi: 10.1016/j.jnutbio.2006.12.011. [DOI] [PubMed] [Google Scholar]
  • 210.Perez-Bosque A, Miro L, Polo J, Russell L, Campbell J, et al. Dietary plasma proteins modulate the immune response of diffuse gut-associated lymphoid tissue in rats challenged with Staphylococcus aureus enterotoxin B. J Nutr. 2008;138:533–37. doi: 10.1093/jn/138.3.533. [DOI] [PubMed] [Google Scholar]
  • 211.Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, et al. IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat Med. 2008;14:275–81. doi: 10.1038/nm1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Investig. 2008;118:534–44. doi: 10.1172/JCI33194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Radaeva S, Sun R, Pan HN, Hong F, Gao B. Interleukin 22 (IL-22) plays a protective role in T cell-mediated murine hepatitis: IL-22 is a survival factor for hepatocytes via STAT3 activation. Hepatology. 2004;39:1332–42. doi: 10.1002/hep.20184. [DOI] [PubMed] [Google Scholar]
  • 214.Brubaker PL, Anini Y. Direct and indirect mechanisms regulating secretion of glucagon-like peptide-1 and glucagon-like peptide-2. Can J Physiol Pharmacol. 2003;81:1005–12. doi: 10.1139/y03-107. [DOI] [PubMed] [Google Scholar]
  • 215.Yazbeck R, Howarth GS, Abbott CA. Growth factor based therapies and intestinal disease: Is glucagon-like peptide-2 the new way forward? Cytokine Growth Factor Rev. 2009;20:175–84. doi: 10.1016/j.cytogfr.2009.02.008. [DOI] [PubMed] [Google Scholar]
  • 216.Buchman AL, Katz S, Fang JC, Bernstein CN, Abou-Assi SG. Teduglutide, a novel mucosally active analog of glucagon-like peptide-2 (GLP-2) for the treatment of moderate to severe Crohn’s disease. Inflamm Bowel Dis. 2010;16:962–73. doi: 10.1002/ibd.21117. [DOI] [PubMed] [Google Scholar]

RESOURCES