GE48CH16-Finlay

ARI

ANNUAL REVIEWS

13 October 2014

6:23

Further

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Click here for quick links to Annual Reviews content online, including: • Other articles in this volume • Top cited articles • Top downloaded articles • Our comprehensive search

Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens Sophie Yurist-Doutsch,1 Marie-Claire Arrieta,1 Stefanie L. Vogt,1 and B. Brett Finlay1,2,3 1 Michael Smith Laboratories, The University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z4; email: [email protected], [email protected], [email protected], bfi[email protected] 2 Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z4 3 Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada, V6T 1Z4

Annu. Rev. Genet. 2014. 48:361–82

Keywords

First published online as a Review in Advance on September 10, 2014

intestinal microbiota, colonization resistance, Salmonella enterica, Escherichia coli, Clostridium difficile

The Annual Review of Genetics is online at genet.annualreviews.org This article’s doi: 10.1146/annurev-genet-120213-092421 c 2014 by Annual Reviews. Copyright  All rights reserved

Abstract The gastrointestinal (GI) microbiota is a complex community of microorganisms residing within the mammalian gastrointestinal tract. The GI microbiota is vital to the development of the host immune system and plays a crucial role in human health and disease. The composition of the GI microbiota differs immensely among individuals yet specific shifts in composition and diversity have been linked to inflammatory bowel disease, obesity, atopy, and susceptibility to infection. In this review, we describe the GI microbiota and its role in enteric diseases caused by pathogenic Escherichia coli, Salmonella enterica, and Clostridium difficile. We discuss the central role of the GI microbiota in protective immunity, resistance to enteric pathogens, and resolution of enteric colitis.

361

GE48CH16-Finlay

ARI

13 October 2014

6:23

THE GASTROINTESTINAL MICROBIOTA: DEFINITION AND COMPOSITION

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

The human gastrointestinal tract (GIT) is home to a complex microbial community with as many as 100 trillion prokaryotic members (119). Although mammals are colonized by many single-cell organisms at various anatomical locations, the GIT harbors the highest number and density of bacteria (67). This complex ecosystem is initially acquired during and for some time after birth. Although the GIT composition varies during the first year of life, at around one year of age the GIT microbiota of an individual becomes stable (87) and remains largely unchanged unless perturbed by antimicrobial treatment or disease (18). The composition of the gastrointestinal (GI) microbiota is highly variable among individuals. Analysis of stool samples collected from healthy subjects showed that the vast majority of bacteria in the GIT belong to only four phyla: Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria (27, 30, 38, 65). At the genus level, the diversity is so high that no two stool samples (of more than 250 tested samples in the Human Microbiome Project) have the same composition (38). Within the GIT, microbial communities differ in the stomach, small intestine, cecum, and colon, with higher levels of bacterial diversity in the lower segments of the GIT and higher levels of aerobic and facultative anaerobic bacteria in the stomach and upper small intestine (30, 35). Additionally, the bacteria present in stool differ from those that are adherent to the GI mucosa (27). Despite the high variability in GIT microbiota composition among individuals, metagenomic analysis reveals high levels of similarity between subjects when comparing various metabolic pathways within microbiota genes (38, 57), suggesting a high level of metabolic redundancy among different bacterial taxa.

IMPACT OF THE GASTROINTESTINAL TRACT MICROBIOTA ON THE IMMUNE SYSTEM The Mammalian Immune System The immune system is defined as the host’s defense against destructive forces from both outside (e.g., bacteria, viruses, parasites) and within (e.g., malignant and autoreactive cells) the body. Immune responses are generally classified as either innate or acquired. The components and cells that make up these two arms of the immune system are presented in Table 1. The innate immune system provides immunity to invading organisms without the need for prior exposure to these antigens and includes physical barriers, such as the skin and mucous membranes; cell-mediated barriers, including phagocytic cells, inflammatory cells, dendritic cells, and natural killer cells; and soluble mediators, such as cytokines, complement proteins, and acutephase proteins (24). This arm of the immune system provides the early phases of host defense that protect the organism during the four to five days it takes for lymphocytes to become activated. The acquired, or adaptive, immune system develops over an individual’s lifetime. Lymphocytes are an important cellular component of this arm of the immune system that modulate the function of other immune cells or directly destroy cells infected with intracellular pathogens (Table 1). Each developing T or B cell generates a unique receptor, or recognition molecule, such that a set of cells expressing a vast array of diverse receptors is produced, allowing immune cells to selectively eliminate virtually any foreign antigen that enters the body (24). B cells, abundant in lymph nodes, recognize foreign antigen through membrane-bound antibodies, or immunoglobulins, and upon activation become antibody-secreting plasma cells to effectively remove soluble bacteria/antigens (24). 362

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

13 October 2014

6:23

Table 1 The immune system Arm of immune system

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Innate immune system

Acquired immune system

Defenses

Components

Functions

Physical barriers

Skin Mucous membranes

Prevent the entry of antigens into systemic circulation

Cell-mediated barriers

Phagocytic cells, e.g., neutrophils, macrophages

Engulf foreign antigens

Inflammatory cells, e.g., basophils, mast cells Natural killer cells Dendritic cells

Release inflammatory mediators, e.g., histamine, prostaglandins

Soluble factors

Cytokines Complement proteins Acute-phase proteins

Destroy infected or malignant cells Present antigens to lymphocytes

B lymphocytes T lymphocytes

Plasma cells

Activate/recruit other cells Enhance phagocytosis Promote repair of damaged tissue

CD4+ T cells Th1 cells Th2 cells Th17 cells Tregs

Secrete antibodies Induce activation of lymphocytes Promote cell-mediated responses Promote humoral (antibody) responses

CD8+ T cells Cytotoxic T cells Suppressor T cells

Peripheric tolerance Destroy infected or malignant cells Suppress activity of lymphocytes

T cells express a T-cell receptor (TCR) that recognizes a foreign antigen presented in complex with a major histocompatibility complex (MHC) molecule on the surface of an antigen-presenting cell (APC) (24). Subpopulations of T cells include the helper T (Th) cells, which are identified by the presence of the membrane glycoprotein CD4, and cytotoxic T cells that express the CD8 glycoprotein (24). CD4+ cells are classified into a number of Th types: Th1, Th2, Th17, and Treg (16, 24, 68). Viral infections or microbes that infect macrophages or natural killer (NK) cells elicit a Th1 response. Th1 lymphocytes secrete interferon gamma (IFN-γ) and tumor necrosis factor beta (TNF-β). A Th2 response elicits Th2 lymphocytes in response to helminths, allergens, and extracellular microbes. Th2 lymphocytes produce cytokines interleukin (IL)-4, 5, and 13, among others. Th17 cells are lymphocytes that produce IL-17 to recruit neutrophils and macrophages and to trigger an inflammatory response in different organs in order to remove extracellular pathogens from the body. Additionally, there is a regulatory arm characterized by regulatory T cells (Tregs) that produce IL-10 to downregulate all other immune arms with the objective of preventing an exacerbated and injury-provoking immune response. Achieving a balance between all these arms is what enables an adequate immune response to fight pathogens and prevents immune-mediated diseases, such as diabetes type 1, rheumatoid arthritis, inflammatory bowel disease (IBD), allergies, asthma, etc.

The Gut-Associated Lymphoid Tissues The mucosal immune system is strategically located in areas where external pathogens and antigens may gain access to the body. This includes the mucosal-associated lymphoid tissues, which www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

363

GE48CH16-Finlay

ARI

13 October 2014

6:23

Villus

INTESTINAL LUMEN

IgA

Goblet cell

Intraepithelial lymphocyte DC

Bacteria

B cell

Epithelium

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

M cell

Peyer’s patch DC

T cell

GC

Crypt

PC Isolated lymphoid follicle

GC

GC

Macrophage

T cells Mesenteric lymph node

LAMINA PROPRIA

Figure 1 Diagram of the gut-associated lymphoid tissue (GALT). The GALT consists of aggregated lymphoid follicles (Peyer’s patches and isolated lymphoid follicles) and diffuse or nonaggregated tissue (lamina propria). The epithelium of aggregated follicles contains M cells, which are specialized in antigen sampling. Underneath is the subepithelial dome, which consists of germinal centers (GCs) of B cells in different stages of maturation, as well as dendritic cells (DCs) and T cells. Aggregated follicles are the inductive sites of immunity, where na¨ıve T and B cells are activated to then migrate to the mesenteric lymph node and into the effector sites of the GALT, the lamina propria. Within the lamina propria, mature B cells or plasma cells (PCs) produce IgA, which gets actively transported into the intestinal lumen. DCs in the lamina propria also sample antigens and present them directly to T and B cells. Macrophages are important innate immune cells capable of engulfing and killing microorganisms by phagocytosis. Figure adapted from Reference 2.

protect sites such as the respiratory, urinary, and reproductive tracts, and the gut-associated lymphoid tissues (GALTs), which protect the intestine. As the intestine is the first line of defense from the environment and must integrate complex interactions among diet, external pathogens, and local immunological processes, it is critical that protective immune responses are mounted against potential pathogens, yet it is equally important that hypersensitivity reactions to dietary and commensal microbial antigens are minimized. The GALT is composed of aggregated tissue in the form of Peyer’s patches and solitary lymphoid follicles, nonaggregated cells in the lamina propria and intraepithelial regions of the intestine, and mesenteric lymph nodes (58) (Figure 1). Peyer’s patches are aggregates of lymphoid follicles found throughout the mucosa and submucosa of the small intestine. These patches contain both CD4+ and CD8+ T cells, as well as na¨ıve B cells, plasma cells, macrophages, and dendritic cells (58). Overlying the Peyer’s patches are specialized epithelial cells known as M cells, which endocytose, transport, and release antigens from the gut into the Peyer’s patches, where these antigens are presented on APCs to T and B cells (50, 58). The lamina propria harbors a diffuse population of T and B cells, plasma cells, dendritic cells, mast cells, and macrophages, all covered by a single layer of epithelial cells (58). Intestinal 364

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

13 October 2014

6:23

epithelial cells (IECs) or enterocytes are intimately involved in digestion, absorption, and transport of nutrients but also play a major role in immune regulation of the underlying cells of the lamina propria. IECs are known to produce different chemokines and cytokines depending on the type of microbial molecules that come in contact with the epithelium (40). IECs have been shown to condition the immune response of dendritic cells by inducing, through an unknown mechanism, the secretion of tolerogenic signals (IL-10) or active immunity signals (IL-12) by the dendritic cells (91). IECs are also involved in antigen transport to underlying antigen-presenting cells, or they can present the antigen themselves to lymphocytes.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

The Interactions Between the Microbiota and the Gut-Associated Lymphoid Tissues Life without symbiotic microorganisms has a profound effect on the host immune system. Experiments in germ-free mice have consistently shown that many components of the immune system remain underdeveloped until bacterial colonization occurs. Structurally, the aggregated lymphoid structures of the GALT, mesenteric lymph nodes (MLNs), and isolated lymphoid follicles and Peyer’s patches are reduced in size and cellularity in germ-free mice (11, 72). Functionally, these mice are depleted in the production of many cytokines, CD4+ T helper cells, Tregs, B cells, Th17 cells, and antimicrobial peptides, and in expression of the MHC class II, etc. (26, 31, 43, 77, 83, 92, 107). Many of these defects can be reverted upon inoculation with a single species of bacteria (72, 112), indicating that it is the interactions with the microbiota that kick-start the postnatal phase of immune development. Gut bacteria also have critical effects on the development of IECs. IECs express microbial pattern recognition receptors (PRRs) that bind to microbial signals and upregulate the expression of mucus, cytokines, and other immune components (90). In axenic mice, IECs have blunted microvilli, and they display reduced expression of PRRs (1, 122). Colonization of the host GIT by the microbiota produces inflammatory and tolerogenic signals that stimulate and regulate the different arms of the immune response in order to achieve homeostasis, a state that is favorable to both the microbiota and the host. The Th17 immune response provides a good example of these opposing functions. Despite the fact that Th17 cells are critical in controlling extracellular bacterial and fungal infections (100, 126), overproduction of Th17 cytokines IL-17 and IL-23 is associated with colitis (3, 13) and autoimmunity (64, 118, 126). Thus, the appropriate level of the Th17 response must be achieved to prevent microbial attack and avoid uncontrolled inflammation. An example of a member of the microbiota involved in regulating the Th17 response has been described in segmented filamentous bacteria (SFB), recently named Candidatus Savagella. SFB are a clostridial species that resides in the murine ileum and are a potent stimulator of the Th17 response (31, 126). Colonization with SFB induced the upregulation of cytokines, antimicrobial peptides, and serum amyloid A (SAA), an acute-phase protein secreted during inflammation. SAA is believed to promote the Th17 differentiation of CD4+ T cells (5, 42). Not surprisingly, microbial signals have also been found to limit the Th17 immune response. Unspecified commensal bacteria induce IECs to produce IL-25, which inhibits IL-23 production by dendritic cells, thus preventing Th17 differentiation (128). In addition, polysaccharide A (PSA) from Bacteroides fragilis downregulated the Th17 response and induced propagation of Tregs in mice that are genetically predisposed to develop autoimmune encephalomyelitis. This treatment was sufficient to prevent disease in this animal model (85). These examples show how microbial signals can act as the tipping forces that increase or decrease Th17 immune cell activation. www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

365

ARI

13 October 2014

6:23

Although certain microbes induce a proinflammatory response and others a tolerogenic one, how the immune response differentiates between them is unknown. One way that microbiota may induce a tolerogenic immune response is via their interactions with Tregs. Tregs have a central role in regulating inflammatory responses, and they have been involved in the modulation of disease in animal models of asthma (94) and colitis (4) upon changes in the intestinal microbiome. Although a large proportion of Tregs are developed in the thymus with the purpose of generating tolerance toward self-antigens and preventing autoimmune responses, a subset of Tregs are trained by the GIT microbiota. Analysis of several repertoires of the TCR α chain revealed that the TCRs from Treg cells of the colonic lamina propria are highly heterogeneous compared with Treg cells from other lymphoid organs. Moreover, the study showed that the mouse microbiota was essential for the induction of this particular population of colonic Treg cells from na¨ıve T cells, implying that there may be post-thymic mechanisms of immune cell education that occur via interactions with the commensal microbiota (60). Thus, besides protecting the host from microbial attack, postnatal bacterial colonization may serve the immune system with another important function: to be used as a training ground for a specific population of immune cells.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

THE ROLE OF THE MICROBIOTA IN ENTERIC INFECTION Although the GIT microbiota is often referred to as commensal, with the implication that these organisms are neither harmful nor beneficial to their host, numerous studies clearly demonstrate that the composition and diversity of the GIT microbiota are crucial to host health and disease. Increasing numbers of studies provide evidence for the importance of the GIT microbiota in many human disorders, including immune-mediated and metabolic diseases such as inflammatory bowel disease (30, 54, 70, 120), obesity (65, 110, 129), asthma, atopy (10, 51, 93), etc. In the following sections, we focus on the key role of the GIT microbiota in the susceptibility to and recovery from enteric infection.

Colonization Resistance The microbiota can determine host susceptibility to bacterial infections. Germ-free and antibiotictreated mice are significantly more susceptible to infectious disease caused by intestinal pathogens, including Shigella flexneri (103), Citrobacter rodentium (125), Listeria monocytogenes (127), and Salmonella enterica (29, 81, 99). The microbiota can prevent or ameliorate infection by direct microbial antagonism or by indirectly promoting appropriate host immune defenses. Direct microbial effects, also defined as colonization resistance (114), include competition for nutrients and host receptors and secretion of antimicrobial substances (97). For example, the probiotic strain Escherichia coli Nissle 1917 reduces Salmonella Typhimurium colonization of the mouse intestine by competing for iron, a limiting nutrient in vivo (25). As an example of indirect effects via immune modulation, administration of the Toll-like receptor (TLR) 5 ligand flagellin to antibiotic-treated mice inhibited colonization with vancomycin-resistant Enterococcus (VRE) by upregulating the secretion of the bactericidal lectin REGIIIγ in IECs and Paneth cells (56). Flagellin stimulation has also been shown to induce differentiation of nonspecific IgA+ plasma cells via activation of TLR5 in intestinal dendritic cells (111). Colonization with B. fragilis is sufficient to prevent murine experimental colitis induced by Helicobacter hepaticus. B. fragilis is coated with PSA, and this capsular component was shown to suppress IL-17, increase IL-10 production, and ameliorate disease in this model (73). Collectively, these findings suggest that the GI microbiota alter the course of bacterial infections both directly and indirectly via the host’s immune response. In the following 366

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

13 October 2014

6:23

section, we discuss in detail interactions between the GIT microbiota and three major enteric pathogens: E. coli, S. Typhimurium, and Clostridium difficile.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Attaching/Effacing Pathogens: Enterohemorrhagic Escherichia coli–Enteropathogenic Escherichia coli–Citrobacter rodentium E. coli is a common commensal inhabitant of the mammalian intestinal tract that can become a GI or extraintestinal pathogen by genetically acquiring pathogenicity islands and plasmids (21). Among the several E. coli pathotypes that cause enteric infections, two of the best studied are enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC). Both EPEC and EHEC are attaching and effacing (A/E) pathogens, which are characterized by their ability to adhere tightly to the host intestinal epithelium and cause localized destruction (effacement) of microvilli (20, 53). These phenotypes are conferred by the chromosomal pathogenicity island known as the locus of enterocyte effacement (LEE), which encodes a type III secretion system (T3SS) and a variety of effector proteins that are secreted into host cells, leading to rearrangement of the actin cytoskeleton (95). Additionally, EHEC but not EPEC carries the stx genes, encoding the Shiga toxin that is responsible for the more severe outcomes of EHEC infection, such as hemolytic uremic syndrome (21). Because EPEC and EHEC are human-specific pathogens that do not cause representative infections in model hosts such as mice (78), C. rodentium, which carries the LEE and is a natural pathogen of mice, is frequently used as a surrogate to study these host-pathogen and microbiota-pathogen interactions and mechanisms (79). C. rodentium infections of mice have provided convincing evidence that the microbiota plays a critical role in determining the outcome of infection with A/E pathogens. As one extreme example, germ-free mice have an impaired ability to clear C. rodentium infections. Specific pathogen-free C57BL/6 mice, possessing an intact microbiota, clear C. rodentium infections within 22 days, whereas germ-free mice of the same strain remain colonized with C. rodentium even 42 days post infection (dpi) (52). Less drastic perturbations of the microbiota, such as treatment with antibiotics, can also affect the outcome of infection. Treating mice with metronidazole prior to C. rodentium infection leads to more severe cecal inflammation 6 dpi (125). Notably, the overall abundance of gut microbiota was not altered by the metronidazole treatment in this study, suggesting that specific microbes whose abundance was altered by metronidazole play an important role in protecting or predisposing the host to C. rodentium–induced colitis (125). Even differences in microbiota composition between strains of mice can have drastic effects on C. rodentium infection. Mouse strains have long been known to vary in their susceptibility to C. rodentium: Resistant strains, such as NIH Swiss and C57BL/6, survive and clear the infection, whereas C. rodentium infection is lethal to susceptible strains, such as C3H/HeJ (113). In order to assess whether differences in gut microbiota composition in these mouse strains are responsible for the different infection outcomes, microbiota (fecal) transplantation was performed. In these experiments, recipient mice were first treated with antibiotics to deplete their natural microbiota, then repeatedly gavaged with donor fecal extracts from either the same or a different strain of mice to establish a new gut microbial community. Remarkably, transplantation of resistant microbiota into a susceptible recipient significantly increased survival of C. rodentium infection compared with mock-treated susceptible mice (33, 121). There are several mechanisms by which the microbiota can affect the ability of A/E pathogens to infect their hosts (Figure 2a). One of these mechanisms is competition for nutrients. Other enterobacteria, particularly commensal E. coli, are especially effective at competing for nutrients with A/E pathogens. Indeed, precolonization of mice with three commensal strains of E. coli prevents subsequent colonization by EHEC (63). There are likely several limiting nutrients for which www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

367

GE48CH16-Finlay

ARI

13 October 2014

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Figure 2 The gastrointestinal (GI) microbiota during Citrobacter rodentium infection in mice. (a) Early stage of infection: C. rodentium enters the GI tract (GIT), where short-chain fatty acids (SCFAs) (as well as other potential mechanisms) lead to an increase in T3SS (type III secretion system) expression and attachment to the epithelium. Variation in GI microbiota composition between mice strains results in varying susceptibility to the pathogen. A GI microbiota poor in commensal Escherichia coli allows superior pathogen proliferation in the GIT. (b) Late stage of infection: Two populations of C. rodentium are present in the GIT. One is attached to the epithelium and the other replicates in the lumen. Microbiotamediated increase in mucin secretion by goblet cells helps detach the pathogen and push it out of the GIT. (c) End stage of infection: The regeneration of the GIT microbiota leads to effective pathogen exclusion. Abbreviation: A/E, attaching and effacing.

368

6:23

a

Early A/E pathogen

Commensal E.. coli

Enterocyte

T3SS

M cell

Goblet cell Dendritic cell Macrophage

b

Neutrophil

Late Segmented filamentous bacteria Microbiota Short-chain fatty acid

c

End

Yurist-Doutsch et al.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

ARI

13 October 2014

6:23

EHEC and commensal enterobacteria compete, including proline and mono- and disaccharides (47, 76). The importance of competition for sugars is supported by studies of germ-free mice monocolonized with C. rodentium (52). When these mice are subsequently colonized with commensal E. coli, which like C. rodentium preferentially metabolizes monosaccharides, the intestinal load of C. rodentium is reduced by several orders of magnitude. However, colonization with Bacteroides thetaiotaomicron, which can metabolize polysaccharides that are not used by C. rodentium, fails to decrease C. rodentium colonization unless mice are fed on a simple sugar diet that forces the bacteria to compete for monosaccharides (52). Colonization of the epithelial surface, rather than the lumen of the intestine or the outer mucus layer where the microbiota reside (45), may be one strategy that A/E pathogens employ to reduce competition for nutrients. C. rodentium T3S mutants that cannot adhere to the host epithelium can colonize germ-free mice but are unable to establish an infection in mice with an intact microbiota (52). Association with the epithelium may allow A/E pathogens to access nutrients that are not available to microbiota in the lumen of the intestine, such as the products of plasma membrane damage (75). The GIT microbiota also exerts indirect effects on A/E pathogens via the host immune system (Figure 2b). The barrier posed by the mucus layer is a central part of the defense against A/E pathogens (and most other enteric pathogens). Mucus secretion from goblet cells in the colon is enhanced during the clearance phase of C. rodentium infections, and Muc2−/− mice that lack the major intestinal mucin protein have a higher rate of mortality during C. rodentium infection than wild-type mice (8, 36). The microbiota appear to play an important role in the maintenance of the mucus layer, as mice treated with the antibiotic metronidazole have a thinner inner mucus layer and are more susceptible to C. rodentium–induced colitis (125). The microbiota from resistant mouse strains also promotes the production of proinflammatory cytokines, such as TNF-α and MIP-2α (33, 121). These cytokines enhance host defenses against C. rodentium by increasing the production of antimicrobial peptides Reg3β and Reg3γ and by promoting infiltration of neutrophils into the colonic mucosa and submucosa (33, 121). IL-22 appears to be particularly important for protection against C. rodentium because treatment of susceptible mice with anti-IL-22 antibodies abrogates the protection normally provided by transplantation with resistant microbiota (121). In addition to changing the ability of A/E pathogens to survive in the GIT by competition and modulation of host immunity, the microbiota also releases metabolites that alter A/E pathogen gene expression. Short-chain fatty acids (SCFAs) are major microbiota-derived metabolites produced from the anaerobic fermentation of dietary fiber in the cecum and the colon (23). When added to culture media at a concentration similar to that found in the human colon, the SCFAs acetate, propionate, and butyrate activate expression of several EHEC virulence genes, including the LEE-encoded T3S proteins EspB and Tir and the chromosomally encoded adhesin Iha (37, 80). SCFAs, particularly butyrate, produced by intestinal microbiota could therefore be a cue for EHEC to trigger the expression of genes needed for adherence and colonization of the colon. Mucus-derived sugars that are liberated by the microbiota could be another signal that EHEC uses to sense its location. Fucose, which is cleaved from intestinal mucin by species such as B. thetaiotaomicron, represses expression of the LEE via the two-component system FusKR (86). Because expression of the LEE represents a significant metabolic burden, this fucose-mediated repression could help EHEC to conserve energy while it is present in the mucus layer rather than in close proximity to the epithelium where the T3SS is needed (86). In addition to these examples, there remain additional uncharacterized metabolites produced by the microbiota that affect EHEC virulence gene expression. For example, growth of EHEC either in a human microbiota–conditioned medium or in the presence of probiotic Lactobacillus or Bifidobacterium species represses the expression of the Stx2 Shiga toxin (14, 23). Further studies are needed to reveal the full scope of how the microbiota affects infections by A/E pathogens. www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

369

GE48CH16-Finlay

ARI

13 October 2014

6:23

Salmonella enterica

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Unlike the A/E pathogens that generally remain in the intestinal lumen, S. enterica are a group of facultative intracellular pathogens that can invade the host tissue as well as proliferate in the gut and as such can generate illnesses ranging from mild gastroenteritis to acute systemic colonization, also known as typhoid fever. Once the pathogen reaches the host GIT, it uses a T3SS encoded by Salmonella pathogenicity island 1 (SPI-1) to invade M cells in Peyer’s patches along the small intestine (46). Once internalized, a second T3SS encoded by Salmonella pathogenicity island 2 (SPI-2) is activated by the pathogen, allowing it to cross the epithelial barrier. In the lamina propria, S. enterica is internalized by macrophages that transfer the pathogen to the internal organs (22). Moreover, S. enterica can be sampled from the intestine by dendritic cells. These phagocytic cells internalize the pathogen directly from the gut lumen and are hijacked by S. enterica for systemic transfer and colonization (115) (Figure 3a). Different serovars of S. enterica are linked to different manifestations of disease. In typhoidal salmonellosis, caused in humans by serovars Typhi and Paratyphi, the pathogen invades the internal organs without generating intestinal inflammation or significant colonization of the GIT. Non-typhoidal salmonellosis results in bacterial GI colonization accompanied by inflammation and pathology, but there is little to no systemic infiltration of the pathogen. S. enterica serovar Typhimurium (S. Typhimurium), an agent of gastroenteritis in humans, can be used to model both typhoidal and non-typhoidal salmonellosis in mice; the shift between the two diseases is moderated by the GIT microbiota. Oral inoculation of C57B/6 mice with S. Typhimurium results in high levels of colonization of the liver, spleen, and mesenteric lymph nodes but not intestinal colonization or pathology. However, if mice are treated 24 hours before infection with a high dose of the antibiotic streptomycin (20 mg), the animals develop GI colonization and inflammation (6). Low doses of antibiotic treatment (streptomycin, vancomycin, or metronidazole at doses of 150–600 mg/L of drinking water) also result in increased susceptibility to S. Typhimurium gastroenteritis, accompanied by GI pathology and proinflammatory cytokine secretion (TNF-α, MCP-1, KC, IL-6) (29, 99). The low-dose streptomycin treatment alters the GIT microbiota composition and reduces microbial diversity but does not significantly reduce the total bacterial count in the gut (29, 32, 99). Similar to the case with C. rodentium, these data suggest that it is not the overall abundance of GIT microbiota but rather their diversity or the presence or absence of certain organisms that determines susceptibility to S. Typhimurium gastroenteritis. Antibiotic treatment also affects S. Typhimurium colonization of other mouse strains, such as 129 SvJ. Unlike the C57B/6 mice that succumb to S. Typhimurium infection within several days, the 129 SvJ mice are more resistant because of host genetic differences and can recover and clear the pathogen. Following S. Typhimurium −−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−→ Figure 3 The gastrointestinal (GI) microbiota during Salmonella enterica serovar Typhimurium gastroenteritis in mice. (a) Early stage of infection: S. Typhimurium invades M cells in Peyer’s patches or is taken up by dendritic cells. The initial composition of the GI microbiota is critical for S. Typhimurium proliferation in the GI tract (GIT). Only mice pretreated with antibiotics or colonized by a low-complexity microbiota develop gastroenteritis following S. Typhimurium infection. Microbiota-mediated release of fucose provides the pathogen with a unique carbon source in the post-antibiotic-treated GIT. (b) Late stage of infection: S. Typhimurium–induced inflammation leads to the release of thiosulfate and ethanolamine, which are used selectively by the pathogen for growth. Furthermore, neutrophil recruitment to the gut lumen and RegIIIβ further reduce the GI microbiota and thus aid S. Typhimurium colonization of the GIT. (c) End stage of infection: The GI microbiota regenerates and pushes the pathogen out of the host. Abbreviation: T3SS, type III secretion system. 370

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

a

13 October 2014

6:23

Early T3SS

S. Typhimurium Enterocyte

M cell

Goblet cell

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Dendritic cell Macrophage

b Late

Neutrophil Segmented filamentous bacteria Microbiota Short-chain fatty acid Thiosulfate Fucose Ethanolamine RegIIIβ

c End

www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

371

ARI

13 October 2014

6:23

infection, up to 25% of infected 129 SvJ mice develop a supershedder phenotype characterized by long-term GI colonization and inflammation as well as continuous fecal shedding of the pathogen. Treatment of the mice with streptomycin or neomycin (5 mg) before infection resulted in all mice having the supershedder phenotype. Even mice that were allowed to recover for seven days after neomycin treatment, a time frame in which the microbiota generally reverts to the pretreated state, remained susceptible to GI colonization, demonstrating the importance of the microbiota for the ability of 129 SvJ mice to clear S. Typhimurium infection (62). Antibiotic treatment was also shown to promote S. Typhimurium GIT colonization in the FvB murine strain (19). In this model, increased susceptibility was observed even after three weeks of post-antibiotic recovery. Perhaps the clearest evidence for the importance of a complex GIT microbiota in preventing/reducing S. Typhimurium colonization was shown using low-complexity microbiota (LCM) mice. These animals are bred germ free and are colonized at birth with the Altered Schaedler Flora, a combination of eight bacterial strains that are representative of the major phylogenetic groups found in conventional murine microbiota. The LCM mice have significant reduction in GI microbial diversity (8 versus >500 strains in conventionally bred mice) but similar bacterial counts in the gut. These animals also develop gastroenteritis when infected with S. Typhimurium without the need for antibiotic pretreatment (104). When housed with conventionally bred animals (which facilitates microbiota transfer between animals), the LCM mice gained partial resistance to S. Typhimurium–induced colitis. The transfer of complex GIT microbiota from the conventionally bred mice to the LCM mice was not complete, and lingering population changes, specifically increased levels of Enterobacteriaceae, were linked with increased susceptibility to S. Typhimurium colitis (104). A simplified model of the post-antibiotic-treated gut was used to understand why it was permissive to S. Typhimurium colonization (82). Germ-free mice were colonized with B. thetaiotaomicron and infected with S. Typhimurium. S. Typhimurium isolated from the gut of these mice was shown to upregulate genes involved in sialic acid and fucose catabolism compared with S. Typhimurium isolated from uncolonized germ-free mice. Furthermore, mutants in these metabolic pathways have reduced virulence in both the B. thetaiotaomicron monocolonized mice and in antibiotictreated animals. Consistently, the GI content of both the B. thetaiotaomicron monocolonized mice and the antibiotic-treated mice contained elevated levels of sialic acid. B. thetaiotaomicron is able to free N-acetylneuraminic acid (Neu5Ac) from intestinal mucin by the action of a specific sialidase. Under normal conditions Neu5Ac is used as a carbon source by various members of the GIT microbiota, however in monocolonized or antibiotic-treated mice the absence of Neu5A metabolizing strains leaves the sugar available for S. Typhimurium consumption. Hydrogen is another metabolite that has been shown to promote S. Typhimurium proliferation in the gut of mice with a simplified microbiota. LCM mice were infected with an S. Typhimurium transposon mutant library, and mutants of the hyb hydrogen utilization operon were found to have attenuated virulence in the model (69). In competition with infections with the wild-type strain, the S. Typhimurium hyb operon mutant displayed attenuated virulence in both LCM and conventional mice. However, in animals treated with a high dose of streptomycin, the hyb mutant had similar fitness to the wild-type strain. Hydrogen is produced by the GIT microbiota during carbon fermentation and was shown in this study to be important in the early stages of S. Typhimurium GIT colonization. Other microbiota-induced metabolic changes in the gut may also affect S. Typhimurium colitis. A directed metabolomics survey of low-dose-streptomycintreated mice found a reduction in SCFA concentration in the ceca (32). The SCFAs propionate and butyrate have been shown to suppress SPI-1 expression in vitro (39, 61), and propionate was also demonstrated to kill S. Typhimurium under gut-like in vitro conditions (17). Collectively,

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

372

Yurist-Doutsch et al.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

ARI

13 October 2014

6:23

these studies strongly support the notion that antibiotic treatment promotes metabolic changes in the GIT that can be conducive to S. Typhimurium colonization (Figure 3a). GI inflammation induced by S. Typhimurium has been generally viewed as a mechanism for pathogen control and expulsion. Recent studies show instead that the pathogen actually manipulates inflammation as a source of nutrients and to out-compete the GIT microbiota in the gut lumen (Figure 2b). In antibiotic-treated animals, wild-type S. Typhimurium induces colitis and inflammation and out-competes the GIT microbiota (98, 105). An S. Typhimurium strain lacking the SPI-1 and SPI-2 secretion systems (the avir strain) can establish initial colonization in the gut of antibiotic-treated mice, but the infection is not sustained and the pathogen is out-competed and expelled by the GIT microbiota. However, in mice that develop spontaneous colitis or if gut inflammation is induced by T-cell transfer, S. Typhimuriumavir can colonize the GIT to the same extent as the wild-type strain (105). This indicates that gut inflammation is actually supporting S. Typhimurium colonization. In the gastroenteritis model, S. Typhimurium infection strongly induces the expression of RegIIIβ, a lectin secreted by the GI mucosa that was also shown to have bacteriocin activity against gram-positive and gram-negative bacteria, including E. coli, but is harmless to S. Typhimurium itself (106). The elevation of RegIIIβ in the intestine was shown to be inflammation dependent, as it does not occur in immune-deficient mice and may be one mechanism by which the pathogen uses the immune response to clear the GIT microbiota. An alternative or parallel mechanism involves S. Typhimurium–induced neutrophil infiltration into the gut lumen. Neutrophil recruitment during S. Typhimurium infection is virulence factor and colitis dependent (98), and elimination of neutrophils by antibody treatment results in a diminished ability of S. Typhimurium to expel the GIT microbiota (34). Furthermore, neutrophil elastase, a serine protease produced by neutrophils during inflammation, was demonstrated to shift the GIT microbiota of mice in a manner that supported S. Typhimurium GIT colonization (34). Neutrophil efflux to the GIT lumen is also accompanied by increased production of reactive oxygen species (ROS) and nitric oxide (NO). Previously believed to be important in controlling pathogens, ROS and NO in fact assist S. Typhimurium in its competition with the GIT microbiota. The GIT microbiota produces H2 S, which is subsequently metabolized into thiosulfate by the gut lumen. Inflammation-induced release of ROS and NO oxidizes thiosulfate into tetrathionate, an electron acceptor used selectively by S. Typhimurium that also limits the ability of other bacteria to replicate (124). Indeed, the ttrA gene responsible for tetrathionate utilization is crucial to the ability of S. Typhimurium to out-compete the GIT microbiota. Tetrathionate respiration in vivo is coupled with the consumption of ethanolamine, a nutrient that is not fermented by the GIT microbiota and as such serves as an abundant carbon source for S. Typhimurium in the GIT (109). The ability of S. Typhimurium to consume ethanolamine was demonstrated to promote colonization of the inflamed GIT but was dependent on the pathogen’s ability to reduce tetrathionate. The picture emerging from all of the above is that the mucosal defense benefits S. Typhimurium by helping the pathogen to out-compete the GIT microbiota. Mice infected with an SPI-2 S. Typhimurium mutant present a self-limiting infection similar to S. Typhimurium gastroenteritis in humans. In this model of the disease, the microbiota begins to regenerate and out-competes the pathogen (Figure 3c). The starting point for microbiota regrowth remains unknown. It may result from a reduction in the availability of the nutrients required by S. Typhimurium for GIT proliferation, such as thiosulfate or sialic acid, but may also be the result of other, yet undiscovered signals. What is clear is that it is a GIT microbiota–, rather than immune–, dependent process. T- and B-cell-depleted mice can clear S. Typhimurium GIT infection but only if they are colonized with a complex microbiota prior to pathogen inoculation (28). In the model described in this study, all of the mice were treated with high-dose streptomycin

www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

373

GE48CH16-Finlay

ARI

13 October 2014

6:23

prior to S. Typhimurium infection. How exactly the GIT microbiota exerts immune-independent protection on the host even though it is cleared from the GI before infection is just one of the issues that require further study in the field of host-Salmonella-microbiota interactions.

Clostridium difficile

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Unlike S. enterica or pathogenic E. coli, C. difficile is often present in the GIT microbiota of healthy individuals, ranging from 1% to 50% of the population (7, 41, 74). As a minor member of the GIT microbiota, C. difficile has no adverse effects on human health. High colonization by the bacteria, however, leads to a condition known as C. difficile–associated diarrhea (CDAD) and is the main cause of pseudomembranous colitis, a severe and sometimes deadly form of antibioticassociated diarrhea (55). C. difficile is a spore-forming gram-positive bacteria that induces severe colitis through the action of two unique toxins: Toxin A and Toxin B (117). Both toxins are glucosyltransferases that are delivered into host cells, where they modify Rho and Ras family GTPase and induce epithelial cell necrosis (44, 48, 49). Furthermore, these toxins have been shown to disrupt epithelial barrier function by modifying tight junction protein interaction (84). It remains unclear whether patients who develop CDAD acquire C. difficile from the environment or from overgrowth of indigenous bacteria (123). It is, however, remarkably clear that the GIT microbiota provides the host with a high level of resistance to this disease. CDAD in adults is almost exclusively found in antibiotic-treated or otherwise susceptible hospitalized patients (88). CDAD is more common in infants under one year of age (59), even without additional risk factors, but children of that age have been reported to harbor an unstable GIT microbiota (87). Furthermore, the vast majority of animal models of C. difficile colonization involve either germ-free mice or various animals that require aggressive antimicrobial therapy prior to inoculation with the pathogen (9) (Figure 4a). Similar to S. Typhimurium– and C. rodentium–induced colitis, increased susceptibility to CDAD can be linked with population shifts of the GIT microbiota rather than complete microbial depletion. Stool samples of hospitalized CDAD patients that were collected prior to the onset of disease harbored a microbial population of reduced diversity with diminished proportions of the phylum Bacteroidetes and families Bacteroidaceae and Clostridiales as well as increased levels of the family Enterococcaceae (116). Additionally, a study that sampled the GIT microbiota of patients suffering from CDAD found that those who experience repeated infection have a higher level of C. difficile colonization of the gut and an overall reduction in microbiota diversity when compared with healthy subjects as well as with patients suffering an initial episode of the disease (15). In an animal study, mice treated with a single dose of clindamycin became susceptible to C. difficile colitis (12). Similar to the effect of low-dose streptomycin treatment, clindamycin administration did not reduce overall bacterial counts in the gut but did significantly lower population diversity. The post-treatment microbial population was dominated by Enterobacteriaceae species and largely reduced in Lachnospiraceae and Barnesiella populations. A key question in the study of CDAD is how antimicrobial treatment and the subsequent change in the GIT microbiota break the natural resistance to C. difficile over-colonization. Ng et al. (82) reported that sialic acid metabolism by C. difficile is critical for colonization of antibiotic-treated mice as well as germ-free mice monocolonized with B. thetaiotaomicron (82). In both murine populations, sialic acid is more available than in conventionally colonized untreated animals. A more recent study employed metabolomics to survey the C. difficile susceptible GIT (108). Significant alterations in many metabolites were observed in mice treated with cefoperazonein in comparison to untreated animals or those that were allowed to recover from the treatment to the point at which they were once again resistant to C. difficile. The bile acid taurocholate was among 374

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

a

13 October 2014

6:23

Figure 4

Early C. difficile Enterocyte

M cell

Goblet cell

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Dendritic cell Macrophage

b Late

Neutrophil Segmented filamentous bacteria Microbiota Antibiotic Sialic acid Toxin A Toxin B

c End

The gastrointestinal (GI) microbiota during Clostridium difficile infection. (a) Early stage of infection: Antibiotic treatment increases susceptibility to C. difficile colonization, possibly by reducing concentrations of microbiota-produced factors that repress C. difficile spore germination. Sialic acid, released by the GI microbiota in the antibiotic-treated GI tract (GIT), is used by the pathogen as an energy source. (b) Late stage of infection: C. difficile release of Toxin A and Toxin B leads to epithelial necrosis and tight junction disruption. There is an increase in concentrations of C. difficile spore germination– promoting molecules. (c) End stage of infection: Transfer of healthy microbiota (fecal transplant) and restoration of gut homeostasis are currently the best treatments for C. difficile infection.

Fecal transfer

www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

375

ARI

13 October 2014

6:23

the metabolites significantly increased in the cecum of susceptible mice. In vitro assays confirmed that taurocholate is an inducer of C. difficile spore germination. This is consistent with previous in vitro studies that reported primary and secondary bile acids as activators and inhibitors of C. difficile spore germination, respectively (101, 102). The metabolomic screen of Theriot et al. (108) also found increases in various sugars in the antibiotic-treated ceca, and the ability of C. difficile to grow on these carbon sources was confirmed. As such, a possible mechanism of C. difficile expansion in the post-antibiotic-treated gut is that increased availability of various nutrients and changes in spore germination signals together lead to increased pathogen proliferation (Figure 4b). The GIT microbiota may also have other means to control C. difficile population levels. Bacillus thuringiensis isolated from stool of a healthy human was shown to produce thuricin CD, a bacteriocin that can kill C. difficile but not other gram-positive bacteria common to the human GIT (89). Although this study does not link the presence or absence of B. thuringiensis to CDAD, it does demonstrate the potential role of direct bacteria-bacteria interactions in controlling pathogens in the gut. Although several antibiotics can be used to treat CDAD, recurrent infections are frequent because of C. difficile’s ability to form resistant spores. Approximately 15–30% of CDAD patients treated with broad-spectrum antibiotics, such as vancomycin and metronidazole, experience recurrence of infection after the first episode and repeated recurrence occurs in as many as 60% of patients (96). New narrow-spectrum antibiotics, such as fidaxomicin, are emerging as improved treatments for CDAD infection (66). Nevertheless, fecal transplant, the transfer of an entire gut population directly to the patient GIT, is currently the most efficient therapy for the disease (71) (Figure 4c).

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

CONCLUDING REMARKS The GIT microbiota plays a crucial role during the various stages of bacterial enteric colitis. Although pathogenic E. coli, S. Typhimurium, and C. difficile have very different virulence pathways and, appropriately, unique interactions with the host and its microbiota, commonalities do exist. The susceptibility to all three pathogens can be increased by a single dose of an antibiotic that does not clear the GIT microbiota but rather shifts it to a composition more permissive to pathogen colonization. Furthermore, all three pathogens are able to utilize metabolites made available through alterations to the GIT bacterial population. Finally, recovery from gastric bacterial infection depends on the restoration of a complex microbial flora in the GIT. In recent years, much data have accumulated about the host-mediated interaction between the GIT microbiota and bacterial pathogens. Future efforts will likely turn toward deciphering the direct molecular interaction between the microbes harbored by the human body and those that try to attack it.

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.

ACKNOWLEDGMENTS We would like to thank Nat F. Brown for figure preparation and Eric M. Brown for critical reading of the review. The Finlay lab is funded by grants from the Canadian Institutes of Health Research (CIHR). 376

Yurist-Doutsch et al.

GE48CH16-Finlay

ARI

13 October 2014

6:23

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

LITERATURE CITED 1. Abrams GD, Bauer H, Sprinz H. 1963. Influence of the normal flora on mucosal morphology and cellular renewal in the ileum. A comparison of germ-free and conventional mice. Lab. Invest. 12:355–64 2. Arrieta Mendez M-C. 2011. The role of small intestinal permeability in the pathogenesis of colitis in the interleukin-10 gene deficient mouse. PhD Diss. Univ. Alberta, Edmonton AB, Can. 3. Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, et al. 2008. ATP drives lamina propria T(H)17 cell differentiation. Nature 455:808–12 4. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, et al. 2011. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331:337–41 5. Ather JL, Ckless K, Martin R, Foley KL, Suratt BT, et al. 2011. Serum amyloid A activates the NLRP3 inflammasome and promotes Th17 allergic asthma in mice. J. Immunol. 187:64–73 6. Barthel M, Hapfelmeier S, Quintanilla-Martinez L, Kremer M, Rohde M, et al. 2003. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect. Immun. 71:2839–58 7. Beaugerie L, Flahault A, Barbut F, Atlan P, Lalande V, et al. 2003. Antibiotic-associated diarrhoea and Clostridium difficile in the community. Aliment. Pharmacol. Thera. 17:905–12 8. Bergstrom KSB, Kissoon-Singh V, Gibson DL, Ma C, Montero M, et al. 2010. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLOS Pathog. 6:e1000902 9. Best EL, Freeman J, Wilcox MH. 2012. Models for the study of Clostridium difficile infection. Gut Microbes 3:145–67 10. Bisgaard H, Li N, Bonnelykke K, Chawes BL, Skov T, et al. 2011. Reduced diversity of the intestinal microbiota during infancy is associated with increased risk of allergic disease at school age. J. Allergy Clinic. Immunol. 128:646–52; e1–5 11. Bouskra D, Brezillon C, Berard M, Werts C, Varona R, et al. 2008. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 456:507–10 12. Buffie CG, Jarchum I, Equinda M, Lipuma L, Gobourne A, et al. 2012. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile– induced colitis. Infect. Immun. 80:62–73 13. Buonocore S, Ahern PP, Uhlig HH, Ivanov II, Littman DR, et al. 2010. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 464:1371–75 14. Carey CM, Kostrzynska M, Ojha S, Thompson S. 2008. The effect of probiotics and organic acids on Shiga-toxin 2 gene expression in enterohemorrhagic Escherichia coli O157:H7. J. Microbiol. Methods 73:125–32 15. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, et al. 2008. Decreased diversity of the fecal microbiome in recurrent Clostridium difficile–associated diarrhea. J. Infect. Dis. 197:435–38 16. Chen Z, Cobbold SP, Waldmann H, Metcalfe SM. 1994. Tolerance induction in concordant heartxenografted mice by CD4 and CD8 monoclonal antibodies. Transplant. Proc. 26:1199–200 17. Cherrington CA, Hinton M, Pearson GR, Chopra I. 1991. Short-chain organic acids at ph 5.0 kill Escherichia coli and Salmonella spp. without causing membrane perturbation. J. Appl. Bacteriol. 70:161–65 18. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. 2009. Bacterial community variation in human body habitats across space and time. Science 326:1694–97 19. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH. 2009. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect. Immun. 77:2741–53 20. Croxen MA, Finlay BB. 2010. Molecular mechanisms of Escherichia coli pathogenicity. Nat. Rev. Microbiol. 8:26–38 21. Croxen MA, Law RJ, Scholz R, Keeney KM, Wlodarska M, Finlay BB. 2013. Recent advances in understanding enteric pathogenic Escherichia coli. Clin. Microbiol. Rev. 26:822–80 22. de Jong HK, Parry CM, van der Poll T, Wiersinga WJ. 2012. Host-pathogen interaction in invasive salmonellosis. PLOS Pathog. 8:e1002933 23. de Sablet T, Chassard C, Bernalier-Donadille A, Vareille M, Gobert AP, Martin C. 2009. Human microbiota-secreted factors inhibit Shiga toxin synthesis by enterohemorrhagic Escherichia coli O157:H7. Infect. Immun. 77:783–90 www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

377

ARI

13 October 2014

6:23

24. Delves PJ, Roitt IM. 2000. The immune system. First of two parts. New Engl. J. Med. 343:37–49 25. Deriu E, Liu JZ, Pezeshki M, Edwards RA, Ochoa RJ, et al. 2013. Probiotic bacteria reduce Salmonella typhimurium intestinal colonization by competing for iron. Cell Host Microbe 14:26–37 26. Dobber R, Hertogh-Huijbregts A, Rozing J, Bottomly K, Nagelkerken L. 1992. The involvement of the intestinal microflora in the expansion of CD4+ T cells with a naive phenotype in the periphery. Dev. Immunol. 2:141–50 27. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, et al. 2005. Diversity of the human intestinal microbial flora. Science 308:1635–38 28. Endt K, Stecher B, Chaffron S, Slack E, Tchitchek N, et al. 2010. The microbiota mediates pathogen clearance from the gut lumen after non-typhoidal Salmonella diarrhea. PLOS Pathog. 6:e1001097 29. Ferreira RB, Gill N, Willing BP, Antunes LC, Russell SL, et al. 2011. The intestinal microbiota plays a role in Salmonella-induced colitis independent of pathogen colonization. PLOS ONE 6:e20338 30. Frank DN, St. Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. 2007. Molecularphylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 104:13780–85 31. Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, et al. 2009. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31:677–89 32. Garner CD, Antonopoulos DA, Wagner B, Duhamel GE, Keresztes I, et al. 2009. Perturbation of the small intestine microbial ecology by streptomycin alters pathology in a Salmonella enterica serovar typhimurium murine model of infection. Infect. Immun. 77:2691–702 33. Ghosh S, Dai C, Brown K, Rajendiran E, Makarenko S, et al. 2011. Colonic microbiota alters host susceptibility to infectious colitis by modulating inflammation, redox status, and ion transporter gene expression. Am. J. Physiol. Gastrointest. Liver Physiol. 301:G39–49 34. Gill N, Ferreira RB, Antunes LC, Willing BP, Sekirov I, et al. 2012. Neutrophil elastase alters the murine gut microbiota resulting in enhanced Salmonella colonization. PLOS ONE 7:e49646 35. Gu S, Chen D, Zhang JN, Lv X, Wang K, et al. 2013. Bacterial community mapping of the mouse gastrointestinal tract. PLOS ONE 8:e74957 36. Gustafsson JK, Navabi N, Rodriguez-Pineiro AM, Alomran AHA, Premaratne P, et al. 2013. Dynamic ˜ changes in mucus thickness and ion secretion during Citrobacter rodentium infection and clearance. PLOS ONE 8:e84430 37. Herold S, Paton JC, Srimanote P, Paton AW. 2009. Differential effects of short-chain fatty acids and iron on expression of iha in Shiga-toxigenic Escherichia coli. Microbiology 155:3554–63 38. Hum. Microbiome Proj. C. 2012. Structure, function and diversity of the healthy human microbiome. Nature 486:207–14 39. Hung CC, Garner CD, Slauch JM, Dwyer ZW, Lawhon SD, et al. 2013. The intestinal fatty acid propionate inhibits Salmonella invasion through the post-translational control of HilD. Mol. Microbiol. 87:1045–60 40. Hurley BP, McCormick BA. 2004. Intestinal epithelial defense systems protect against bacterial threats. Curr. Gastroenterol. Rep. 6:355–61 41. Iizuka M, Konno S, Itou H, Chihara J, Toyoshima I, et al. 2004. Novel evidence suggesting Clostridium difficile is present in human gut microbiota more frequently than previously suspected. Microbiol. Immunol. 48:889–92 42. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, et al. 2009. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139:485–98 43. Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, et al. 2008. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4:337–49 44. Jank T, Aktories K. 2008. Structure and mode of action of clostridial glucosylating toxins: the ABCD model. Trends Microbiol. 16:222–29 45. Johansson MEV, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC. 2008. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 105:15064–69

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

378

Yurist-Doutsch et al.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

ARI

13 October 2014

6:23

46. Jones BD, Ghori N, Falkow S. 1994. Salmonella typhimurium initiates murine infection by penetrating and destroying the specialized epithelial M cells of the Peyer’s patches. J. Exp. Med. 180:15–23 47. Jones SA, Jorgensen M, Chowdhury FZ, Rodgers R, Hartline J, et al. 2008. Glycogen and maltose utilization by Escherichia coli O157:H7 in the mouse intestine. Infect. Immun. 76:2531–40 48. Just I, Richter HP, Prepens U, von Eichel-Streiber C, Aktories K. 1994. Probing the action of Clostridium difficile toxin B in Xenopus laevis oocytes. J. Cell Sci. 107(Pt. 6):1653–59 49. Just I, Selzer J, Wilm M, von Eichel-Streiber C, Mann M, Aktories K. 1995. Glucosylation of Rho proteins by Clostridium difficile toxin B. Nature 375:500–3 50. Kagnoff MF. 1993. Immunology of the intestinal tract. Gastroenterology 105:1275–80 51. Kalliomaki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. 2001. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J. Allergy Clin. Immunol. 107:129–34 52. Kamada N, Kim Y-G, Sham HP, Vallance BA, Puente JL, et al. 2012. Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. Science 336:1325–29 53. Kaper JB, Nataro JP, Mobley HL. 2004. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2:123–40 54. Kassinen A, Krogius-Kurikka L, Makivuokko H, Rinttila T, Paulin L, et al. 2007. The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology 133:24–33 55. Kelly CP, Pothoulakis C, LaMont JT. 1994. Clostridium difficile colitis. New Engl. J. Med. 330:257–62 56. Kinnebrew MA, Ubeda C, Zenewicz LA, Smith N, Flavell RA, Pamer EG. 2010. Bacterial flagellin stimulates toll-like receptor 5-dependent defense against vancomycin-resistant enterococcus infection. J. Infect. Dis. 201:534–43 57. Kurokawa K, Itoh T, Kuwahara T, Oshima K, Toh H, et al. 2007. Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Res. 14:169–81 58. Langkamp-Henken B, Glezer JA, Kudsk KA. 1992. Immunologic structure and function of the gastrointestinal tract. Nutr. Clin. Pract. 7:100–8 59. Larson HE, Barclay FE, Honour P, Hill ID. 1982. Epidemiology of Clostridium difficile in infants. J. Infect. Dis. 146:727–33 60. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, et al. 2011. Peripheral education of the immune system by colonic commensal microbiota. Nature 478:250–54 61. Lawhon SD, Maurer R, Suyemoto M, Altier C. 2002. Intestinal short-chain fatty acids alter Salmonella typhimurium invasion gene expression and virulence through BarA/SirA. Mol. Microbiol. 46:1451–64 62. Lawley TD, Bouley DM, Hoy YE, Gerke C, Relman DA, Monack DM. 2008. Host transmission of Salmonella enterica serovar Typhimurium is controlled by virulence factors and indigenous intestinal microbiota. Infect. Immun. 76:403–16 63. Leatham MP, Banerjee S, Autieri SM, Mercado-Lubo R, Conway T, Cohen PS. 2009. Precolonized human commensal Escherichia coli strains serve as a barrier to E. coli O157:H7 growth in the streptomycintreated mouse intestine. Infect. Immun. 77:2876–86 64. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. 2010. Microbes and Health Sackler Colloquium: proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 108(Suppl. 1):4615–22 65. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. 2006. Microbial ecology: human gut microbes associated with obesity. Nature 444:1022–23 66. Louie TJ, Cannon K, Byrne B, Emery J, Ward L, et al. 2012. Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI. Clin. Infect. Dis. 55(Suppl. 2):S132–42 67. Luckey TD. 1972. Introduction to intestinal microecology. Am. J. Clin. Nutr. 25:1292–94 68. Macdonald JC, Torriani FJ, Morse LS, Karavellas MP, Reed JB, Freeman WR. 1998. Lack of reactivation of cytomegalovirus (CMV) retinitis after stopping CMV maintenance therapy in AIDS patients with sustained elevations in CD4 T cells in response to highly active antiretroviral therapy. J. Infect. Dis. 177:1182–87 69. Maier L, Vyas R, Cordova CD, Lindsay H, Schmidt TS, et al. 2013. Microbiota-derived hydrogen fuels Salmonella typhimurium invasion of the gut ecosystem. Cell Host Microbe 14:641–51 www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

379

ARI

13 October 2014

6:23

70. Malinen E, Rinttila T, Kajander K, Matto J, Kassinen A, et al. 2005. Analysis of the fecal microbiota of irritable bowel syndrome patients and healthy controls with real-time PCR. Am. J. Gastroenterol. 100:373–82 71. Mattila E, Uusitalo-Seppala R, Wuorela M, Lehtola L, Nurmi H, et al. 2012. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology 142:490–96 72. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. 2005. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122:107–18 73. Mazmanian SK, Round JL, Kasper DL. 2008. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453:620–25 74. McFarland LV, Mulligan ME, Kwok RY, Stamm WE. 1989. Nosocomial acquisition of Clostridium difficile infection. New Engl. J. Med. 320:204–10 75. Miranda RL, Conway T, Leatham MP, Chang DE, Norris WE, et al. 2004. Glycolytic and gluconeogenic growth of Escherichia coli O157:H7 (EDL933) and E. coli K-12 (MG1655) in the mouse intestine. Infect. Immun. 72:1666–76 76. Momose Y, Hirayama K, Itoh K. 2008. Competition for proline between indigenous Escherichia coli and E. coli O157:H7 in gnotobiotic mice associated with infant intestinal microbiota and its contribution to the colonization resistance against E. coli O157:H7. Antonie van Leeuwenhoek 94:165–71 77. Moreau MC, Ducluzeau R, Guy-Grand D, Muller MC. 1978. Increase in the population of duodenal immunoglobulin A plasmocytes in axenic mice associated with different living or dead bacterial strains of intestinal origin. Infect. Immun. 21:532–39 78. Mundy R, Girard F, FitzGerald AJ, Frankel G. 2006. Comparison of colonization dynamics and pathology of mice infected with enteropathogenic Escherichia coli, enterohaemorrhagic E. coli and Citrobacter rodentium. FEMS Microbiol. Lett. 265:126–32 79. Mundy R, MacDonald TT, Dougan G, Frankel G, Wiles S. 2005. Citrobacter rodentium of mice and man. Cell. Microbiol. 7:1697–706 80. Nakanishi N, Tashiro K, Kuhara S, Hayashi T, Sugimoto N, Tobe T. 2009. Regulation of virulence by butyrate sensing in enterohaemorrhagic Escherichia coli. Microbiology 155:521–30 81. Nardi RM, Silva ME, Vieira EC, Bambirra EA, Nicoli JR. 1989. Intragastric infection of germfree and conventional mice with Salmonella typhimurium. Braz. J. Med. Biol. Res. 22:1389–92 82. Ng KM, Ferreyra JA, Higginbottom SK, Lynch JB, Kashyap PC, et al. 2013. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature 502:96–99 83. Niess JH, Leithauser F, Adler G, Reimann J. 2008. Commensal gut flora drives the expansion of proinflammatory CD4 T cells in the colonic lamina propria under normal and inflammatory conditions. J. Immunol. 180:559–68 84. Nusrat A, von Eichel-Streiber C, Turner JR, Verkade P, Madara JL, Parkos CA. 2001. Clostridium difficile toxins disrupt epithelial barrier function by altering membrane microdomain localization of tight junction proteins. Infect. Immun. 69:1329–36 85. Ochoa-Reparaz J, Mielcarz DW, Wang Y, Begum-Haque S, Dasgupta S, et al. 2010. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 3:487–95 86. Pacheco AR, Curtis MM, Ritchie JM, Munera D, Waldor MK, et al. 2012. Fucose sensing regulates bacterial intestinal colonization. Nature 492:113–17 87. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. 2007. Development of the human infant intestinal microbiota. PLOS Biol. 5:e177 88. Pothoulakis C, LaMont JT. 1993. Clostridium difficile colitis and diarrhea. Gastroenterol. Clin. N. Am. 22:623–37 89. Rea MC, Sit CS, Clayton E, O’Connor PM, Whittal RM, et al. 2010. Thuricin CD, a posttranslationally modified bacteriocin with a narrow spectrum of activity against Clostridium difficile. Proc. Natl. Acad. Sci. USA 107:9352–57 90. Rescigno M. 2011. The intestinal epithelial barrier in the control of homeostasis and immunity. Trends Immunol. 32:256–64

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

380

Yurist-Doutsch et al.

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

ARI

13 October 2014

6:23

91. Rimoldi M, Chieppa M, Larghi P, Vulcano M, Allavena P, Rescigno M. 2005. Monocyte-derived dendritic cells activated by bacteria or by bacteria-stimulated epithelial cells are functionally different. Blood 106:2818–26 92. Round JL, Mazmanian SK. 2009. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 9:313–23 93. Russell SL, Finlay BB. 2012. The impact of gut microbes in allergic diseases. Curr. Opin. Gastroenterol. 28:563–69 94. Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, et al. 2012. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 13:440–47 95. Schmidt MA. 2010. LEEways: tales of EPEC, ATEC and EHEC. Cell. Microbiol. 12:1544–52 96. Sears P, Ichikawa Y, Ruiz N, Gorbach S. 2013. Advances in the treatment of Clostridium difficile with fidaxomicin: a narrow spectrum antibiotic. Ann. N. Y. Acad. Sci. 1291:33–41 97. Sekirov I, Finlay BB. 2009. The role of the intestinal microbiota in enteric infection. J. Physiol. 587:4159– 67 98. Sekirov I, Gill N, Jogova M, Tam N, Robertson M, et al. 2010. Salmonella SPI-1-mediated neutrophil recruitment during enteric colitis is associated with reduction and alteration in intestinal microbiota. Gut Microbes 1:30–41 99. Sekirov I, Tam NM, Jogova M, Robertson ML, Li Y, et al. 2008. Antibiotic-induced perturbations of the intestinal microbiota alter host susceptibility to enteric infection. Infect. Immun. 76:4726–36 100. Smith K, McCoy KD, Macpherson AJ. 2007. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin. Immunol. 19:59–69 101. Sorg JA, Sonenshein AL. 2008. Bile salts and glycine as cogerminants for Clostridium difficile spores. J. Bacteriol. 190:2505–12 102. Sorg JA, Sonenshein AL. 2009. Chenodeoxycholate is an inhibitor of Clostridium difficile spore germination. J. Bacteriol. 191:1115–17 103. Sprinz H, Kundel DW, Dammin GJ, Horowitz RE, Schneider H, Formal SB. 1961. The response of the germfree guinea pig to oral bacterial challenge with Escherichia coli and Shigella flexneri. Am. J. Pathol. 39:681–95 104. Stecher B, Chaffron S, Kappeli R, Hapfelmeier S, Freedrich S, et al. 2010. Like will to like: abundances of closely related species can predict susceptibility to intestinal colonization by pathogenic and commensal bacteria. PLOS Pathog. 6:e1000711 105. Stecher B, Robbiani R, Walker AW, Westendorf AM, Barthel M, et al. 2007. Salmonella enterica serovar Typhimurium exploits inflammation to compete with the intestinal microbiota. PLOS Biol. 5:2177–89 106. Stelter C, Kappeli R, Konig C, Krah A, Hardt WD, et al. 2011. Salmonella-induced mucosal lectin RegIIIβ kills competing gut microbiota. PLOS ONE 6:e20749 107. Strauch UG, Obermeier F, Grunwald N, Gurster S, Dunger N, et al. 2005. Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis. Gut 54:1546–52 108. Theriot CM, Koenigsknecht MJ, Carlson PE Jr, Hatton GE, Nelson AM, et al. 2014. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun. 5:3114 109. Thiennimitr P, Winter SE, Winter MG, Xavier MN, Tolstikov V, et al. 2011. Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. Proc. Natl. Acad. Sci. USA 108:17480–85 110. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, et al. 2009. A core gut microbiome in obese and lean twins. Nature 457:480–84 111. Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, et al. 2008. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat. Immunol. 9:769–76 112. Umesaki Y, Okada Y, Matsumoto S, Imaoka A, Setoyama H. 1995. 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. 39:555–62 113. Vallance BA, Deng W, Jacobson K, Finlay BB. 2003. Host susceptibility to the attaching and effacing bacterial pathogen Citrobacter rodentium. Infect. Immun. 71:3443–53 www.annualreviews.org • Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens

381

ARI

13 October 2014

6:23

114. van der Waaij D, Berghuis-de Vries JM, Lekkerkerk-van der Wees JEC. 1971. Colonization resistance of the digestive tract in conventional and antibiotic-treated mice. J. Hyg. 69:405–11 115. Vazquez-Torres A, Jones-Carson J, Baumler AJ, Falkow S, Valdivia R, et al. 1999. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature 401:804–8 116. Vincent C, Stephens DA, Loo VG, Edens TJ, Behr MA, et al. 2013. Reductions in intestinal Clostridiales precede the development of nosocomial Clostridium difficile infection. Microbiome 1:18 117. Voth DE, Ballard JD. 2005. Clostridium difficile toxins: mechanism of action and role in disease. Clin. Microbiol. Rev. 18:247–63 118. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, et al. 2008. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature 455:U1109–10 119. Whitman WB, Coleman DC, Wiebe WJ. 1998. Prokaryotes: the unseen majority. Proc. Natl. Acad. Sci. USA 95:6578–83 120. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, et al. 2010. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 139:1844–54; e1 121. Willing BP, Vacharaksa A, Croxen M, Thanachayanont T, Finlay BB. 2011. Altering host resistance to infections through microbial transplantation. PLOS ONE 6:e26988 122. Willing BP, Van Kessel AG. 2007. Enterocyte proliferation and apoptosis in the caudal small intestine is influenced by the composition of colonizing commensal bacteria in the neonatal gnotobiotic pig. J. Anim. Sci. 85:3256–66 123. Wilson KH. 1993. The microecology of Clostridium difficile. Clin. Infect. Dis. 16(Suppl. 4):S214–18 124. Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL, et al. 2010. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature 467:426–29 125. Wlodarska M, Willing B, Keeney KM, Menendez A, Bergstrom KS, et al. 2011. Antibiotic treatment alters the colonic mucus layer and predisposes the host to exacerbated Citrobacter rodentium–induced colitis. Infect. Immun. 79:1536–45 126. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, et al. 2010. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32:815–27 127. Zachar Z, Savage DC. 1979. Microbial interference and colonization of the murine gastrointestinal tract by Listeria monocytogenes. Infect. Immun. 23:168–74 128. Zaph C, Du Y, Saenz SA, Nair MG, Perrigoue JG, et al. 2008. Commensal-dependent expression of IL-25 regulates the IL-23-IL-17 axis in the intestine. J. Exp. Med. 205:2191–98 129. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, et al. 2009. Human gut microbiota in obesity and after gastric bypass. Proc. Natl. Acad. Sci. USA 106:2365–70

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

GE48CH16-Finlay

382

Yurist-Doutsch et al.

ANNUAL REVIEWS Connect With Our Experts

New From Annual Reviews:

ONLINE NOW!

Annual Review of Cancer Biology

cancerbio.annualreviews.org • Volume 1 • March 2017

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Co-Editors: Tyler Jacks, Massachusetts Institute of Technology Charles L. Sawyers, Memorial Sloan Kettering Cancer Center The Annual Review of Cancer Biology reviews a range of subjects representing important and emerging areas in the field of cancer research. The Annual Review of Cancer Biology includes three broad themes: Cancer Cell Biology, Tumorigenesis and Cancer Progression, and Translational Cancer Science.

TABLE OF CONTENTS FOR VOLUME 1:

• How Tumor Virology Evolved into Cancer Biology and Transformed Oncology, Harold Varmus • The Role of Autophagy in Cancer, Naiara Santana-Codina, Joseph D. Mancias, Alec C. Kimmelman • Cell Cycle–Targeted Cancer Therapies, Charles J. Sherr, Jiri Bartek • Ubiquitin in Cell-Cycle Regulation and Dysregulation in Cancer, Natalie A. Borg, Vishva M. Dixit • The Two Faces of Reactive Oxygen Species in Cancer, Colleen R. Reczek, Navdeep S. Chandel • Analyzing Tumor Metabolism In Vivo, Brandon Faubert, Ralph J. DeBerardinis • Stress-Induced Mutagenesis: Implications in Cancer and Drug Resistance, Devon M. Fitzgerald, P.J. Hastings, Susan M. Rosenberg • Synthetic Lethality in Cancer Therapeutics, Roderick L. Beijersbergen, Lodewyk F.A. Wessels, René Bernards • Noncoding RNAs in Cancer Development, Chao-Po Lin, Lin He • p53: Multiple Facets of a Rubik’s Cube, Yun Zhang, Guillermina Lozano • Resisting Resistance, Ivana Bozic, Martin A. Nowak • Deciphering Genetic Intratumor Heterogeneity and Its Impact on Cancer Evolution, Rachel Rosenthal, Nicholas McGranahan, Javier Herrero, Charles Swanton

• Immune-Suppressing Cellular Elements of the Tumor Microenvironment, Douglas T. Fearon • Overcoming On-Target Resistance to Tyrosine Kinase Inhibitors in Lung Cancer, Ibiayi Dagogo-Jack, Jeffrey A. Engelman, Alice T. Shaw • Apoptosis and Cancer, Anthony Letai • Chemical Carcinogenesis Models of Cancer: Back to the Future, Melissa Q. McCreery, Allan Balmain • Extracellular Matrix Remodeling and Stiffening Modulate Tumor Phenotype and Treatment Response, Jennifer L. Leight, Allison P. Drain, Valerie M. Weaver • Aneuploidy in Cancer: Seq-ing Answers to Old Questions, Kristin A. Knouse, Teresa Davoli, Stephen J. Elledge, Angelika Amon • The Role of Chromatin-Associated Proteins in Cancer, Kristian Helin, Saverio Minucci • Targeted Differentiation Therapy with Mutant IDH Inhibitors: Early Experiences and Parallels with Other Differentiation Agents, Eytan Stein, Katharine Yen • Determinants of Organotropic Metastasis, Heath A. Smith, Yibin Kang • Multiple Roles for the MLL/COMPASS Family in the Epigenetic Regulation of Gene Expression and in Cancer, Joshua J. Meeks, Ali Shilatifard • Chimeric Antigen Receptors: A Paradigm Shift in Immunotherapy, Michel Sadelain

ANNUAL REVIEWS | CONNECT WITH OUR EXPERTS 650.493.4400/800.523.8635 (us/can) www.annualreviews.org | [email protected]

GE48-FrontMatter

ARI

22 October 2014

14:49

Contents

Annual Review of Genetics Volume 48, 2014

L1 Retrotransposons and Somatic Mosaicism in the Brain Sandra R. Richardson, Santiago Morell, and Geoffrey J. Faulkner p p p p p p p p p p p p p p p p p p p p p p p p p 1

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

Factors Underlying Restricted Crossover Localization in Barley Meiosis James D. Higgins, Kim Osman, Gareth H. Jones, and F. Chris H. Franklin p p p p p p p p p p p p29 pENCODE: A Plant Encyclopedia of DNA Elements Amanda K. Lane, Chad E. Niederhuth, Lexiang Ji, and Robert J. Schmitz p p p p p p p p p p p p p p49 Archaeal DNA Replication Lori M. Kelman and Zvi Kelman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p71 Molecular Genetic Dissection of Quantitative Trait Loci Regulating Rice Grain Size Jianru Zuo and Jiayang Li p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p99 Exploring Developmental and Physiological Functions of Fatty Acid and Lipid Variants Through Worm and Fly Genetics Huanhu Zhu and Min Han p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 119 Quality Control and Infiltration of Translation by Amino Acids Outside of the Genetic Code Tammy Bullwinkle, Beth Lazazzera, and Michael Ibba p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 149 Vulnerabilities on the Lagging-Strand Template: Opportunities for Mobile Elements Ashwana D. Fricker and Joseph E. Peters p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 167 Self-Organization of Meiotic Recombination Initiation: General Principles and Molecular Pathways Scott Keeney, Julian Lange, and Neeman Mohibullah p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 187 Cancer: Evolution Within a Lifetime Marco Gerlinger, Nicholas McGranahan, Sally M. Dewhurst, Rebecca A. Burrell, Ian Tomlinson, and Charles Swanton p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 215 Diverse Epigenetic Mechanisms of Human Disease Emily Brookes and Yang Shi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 237 From Egg to Gastrula: How the Cell Cycle Is Remodeled During the Drosophila Mid-Blastula Transition Jeffrey A. Farrell and Patrick H. O’Farrell p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 269 v

GE48-FrontMatter

ARI

22 October 2014

14:49

Cellular and Molecular Mechanisms of Single and Collective Cell Migrations in Drosophila: Themes and Variations Shirin M. Pocha and Denise J. Montell p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 295 The Structure and Regulation of Flagella in Bacillus subtilis Sampriti Mukherjee and Daniel B. Kearns p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 319 Transcription-Associated Mutagenesis Sue Jinks-Robertson and Ashok S. Bhagwat p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 341 Gastrointestinal Microbiota–Mediated Control of Enteric Pathogens Sophie Yurist-Doutsch, Marie-Claire Arrieta, Stefanie L. Vogt, and B. Brett Finlay p p 361

Annu. Rev. Genet. 2014.48:361-382. Downloaded from www.annualreviews.org Access provided by University of Reading on 11/20/17. For personal use only.

The Relations Between Recombination Rate and Patterns of Molecular Variation and Evolution in Drosophila Brian Charlesworth and Jos´e L. Campos p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 383 The Genetics of Neisseria Species Ella Rotman and H. Steven Seifert p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 405 Regulation of Transcription by Long Noncoding RNAs Roberto Bonasio and Ramin Shiekhattar p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 433 Centromeric Heterochromatin: The Primordial Segregation Machine Kerry S. Bloom p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 457 Nonadditive Gene Expression in Polyploids Mi-Jeong Yoo, Xiaoxian Liu, J. Chris Pires, Pamela S. Soltis, and Douglas E. Soltis p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 485 Lineage Sorting in Apes Thomas Mailund, Kasper Munch, and Mikkel Heide Schierup p p p p p p p p p p p p p p p p p p p p p p p p p p 519 Messenger RNA Degradation in Bacterial Cells Monica P. Hui, Patricia L. Foley, and Joel G. Belasco p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 537 Population Genomics of Transposable Elements in Drosophila Maite G. Barr´on, Anna-Sophie Fiston-Lavier, Dmitri A. Petrov, and Josefa Gonz´alez p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 561 Genetic, Epigenetic, and Environmental Contributions to Neural Tube Closure Jonathan J. Wilde, Juliette R. Petersen, and Lee Niswander p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 583 Errata An online log of corrections to Annual Review of Genetics articles may be found at http://www.annualreviews.org/errata/genet

vi

Contents

Gastrointestinal microbiota-mediated control of enteric pathogens.

The gastrointestinal (GI) microbiota is a complex community of microorganisms residing within the mammalian gastrointestinal tract. The GI microbiota ...
1002KB Sizes 0 Downloads 6 Views