3,322
Views
10
CrossRef citations to date
0
Altmetric
Review

The multifaceted virulence of adherent-invasive Escherichia coli

, &
Article: 2172669 | Received 30 Sep 2022, Accepted 16 Jan 2023, Published online: 05 Feb 2023

ABSTRACT

The surge in inflammatory bowel diseases, like Crohn’s disease (CD), is alarming. While the role of the gut microbiome in CD development is unresolved, the frequent isolation of adherent-invasive Escherichia coli (AIEC) strains from patient biopsies, together with their propensity to trigger gut inflammation, underpin the potential role of these bacteria as disease modifiers. In this review, we explore the spectrum of AIEC pathogenesis, including their metabolic versatility in the gut. We describe how AIEC strains hijack the host defense mechanisms to evade immune attrition and promote inflammation. Furthermore, we highlight the key traits that differentiate AIEC from commensal E. coli. Deciphering the main components of AIEC virulence is cardinal to the discovery of the next generation of antimicrobials that can selectively eradicate CD-associated bacteria.

Introduction

The role of the gut microbiome in promoting human health is indubitable. It extends from providing colonization resistance against invading microbes to expanding the digestive ability of the host and regulating its immune functions. In this regard, the metabolites produced by the gut microbiome are known to regulate immune homeostasis and promote mucosal integrity.Citation1 This intertwined connection between gut health and the microbiome motivated the investigation of their role in driving inflammatory bowel diseases like Crohn’s Disease (CD). In addition to negatively impacting the quality of life, CD imposes a significant burden on the healthcare system due to its recurrent fulminant episodes that can lead to repeated hospitalization throughout the lifetime of the patient.Citation2 The increasing incidence of CD in the Western hemisphere since 1960 suggests that the factors predisposing for the disease are surging.Citation3 Genome-wide association studies have identified over 160 susceptible loci that may contribute to the increased risk of developing inflammatory bowel diseases (IBD) like CD.Citation4–7 This includes many loci that are involved in sensing and responding to microbes, suggesting that the microbiome is implicated in CD etiology. In accord with this, the contribution of host genetics to CD development was found to be more prominent in the pediatric population compared to adults, which sheds light on the role of the environmental factors as disease modifiers. Additionally, the concordance of CD among monozygotic twins was found to be approximately 50%,Citation8 confirming that genetic and non-genetic risk factors converge to propel disease development. While CD is marked by a dysregulated immune response to the gut microbiome, the role of specific microbes in promoting these pathological interactions remains unclear.

Due to its contribution to the development of intestinal granulomatous diseases in ruminants, Mycobacterium avium subspecies paratuberculosis (MAP) was proposed to be the causative agent of CD.Citation9–11 Although detected by PCR in the intestinal tissues of CD patients, MAP was also found in the biopsies collected from the control group.Citation12 While these bacteria are commonly associated with livestock, no increase in CD incidence among farmers was reported, a trend that would be expected for a zoonotic pathogen.Citation13 Additionally, anti-Mycobacterial therapies did not improve remission rates among CD patients.Citation14 Other studies employed immunostaining of clinical samples to investigate the potential contribution of bacterial pathogens to CD.Citation15 Using biopsies collected from 21 patients, Liu et al showed a correlation between CD and Escherichia coli, Listeria monocytogenes, and Streptococcus spp. However, another study ruled out a role for Listeria in disease development, since immunostaining revealed no preferential abundance of these bacteria in the CD gut.Citation16 Of note, it is unclear whether the inconsistency between studies in defining the CD-associated microbial community reflects the presence of different subtypes of the disease.

In the quest to isolate the gut microbes that are associated with CD, the late Dr. Darfeuille-Michaud identified a new pathogroup of bacteria called adherent-invasive Escherichia coli (AIEC).Citation17 The likelihood of isolating these bacteria from CD patients is approximately six-times higher than from healthy individuals, suggesting a role in disease development.Citation18,Citation19

Members of the AIEC group can invade the intestinal epithelial cells and replicate inside macrophages similar to entero-invasive E. coli strains. Nevertheless, AIEC strains lack several prominent virulence factors that are known to promote invasion, such as type III secretion system.Citation20,Citation21 Comparative genomics revealed a significant strain divergence among the AIEC group and a lack of a defining genetic blueprint, making this pathovar genetically inextricable from commensal E.coli.Citation20,Citation22,Citation23 While phylogenetic analysis suggests that AIEC strains belong primarily to the B2 clade,Citation20,Citation24–26 a different phylogroup distribution was reported for the strains isolated from the pediatric patient population.Citation27 Interestingly, this remarkable genetic heterogeneity was also observed within the AIEC populations isolated from individual patients.Citation24,Citation27 Similarly, diversification marks the evolutionary response of AIEC to host selection in CD infection models.Citation28

To this end, the identification of AIEC relies on time consuming and non-standardized phenotypic assays in vitro.Citation23 Although the evolutionary origins of AIEC are nebulous, several groups demonstrated that these bacteria could trigger inflammation in the preclinical models of colitis; thus, fulfilling Koch’s postulates in the susceptible hosts.Citation29–32 However, the intricacies of AIEC-host interactions, and how these pathobionts evolved differently from commensals, remain unknown. Unlike commensals, the pathogenic potential of pathobionts manifests under certain host conditions.Citation33 In this article, we review the factors that promote the virulence of AIEC in the host environment. We discuss how AIEC strains can modulate the immune responses to persist in the gut. Additionally, we highlight several examples of the metabolic versatility of this group of bacteria, which is key to their in vivo fitness. While there are other bacteria that have been implicated in CD, their contribution to disease development is beyond the scope of this review.

The main features of AIEC pathogenesis

The ability of AIEC strains to adhere to and invade epithelial cells distinguishes them from commensal E. coli. This is mediated by an arsenal of virulence factors including the adhesin of type I pili, FimH, which can bind to Carcinoembryonic Antigen-related Cell Adhesion Molecules 6 (CEACAM6) receptor.Citation34,Citation35 Interestingly, these receptors are upregulated in the ileal lesions of CD patients, possibly influencing the trajectory of AIEC adaptive evolution.Citation34 This is evident by the enhanced CEACAM6-binding ability of the FimH alleles within the AIEC group relative to their homologues in commensal E. coli.Citation36 While these allelic variations are not always captured by comparative genomics studies, they highlight the different evolutionary trajectories of AIEC and commensals. In line with its role in promoting host colonization, blocking FimH using drugsCitation37 or glycoconjugatesCitation38 curtailed bacterial adhesion to the intestinal epithelium and ameliorated inflammation. In addition to FimH, OmpC was shown to be important for the ability of AIEC to adhere to the intestinal cells. Although its binding target in the host cells remains unknown, OmpC was upregulated in vitro by high osmolarity similar to that encountered by bacteria in the gut, suggesting a role for this protein in promoting in vivo fitness.Citation39 In line with this, antibodies targeting OmpC were significantly more abundant in the sera of CD patients relative to healthy individuals, and could be used in combination with other antibodies as reliable markers for IBD.Citation40 ChiA is another adhesin that mediates the ability of AIEC to attach firmly to the intestinal epithelial cells.Citation41 Similar to other chitin-binding domains in Serratia marcescens and Vibrio cholera,Citation42,Citation43 ChiA binds to Inducible Chitinase 3-Like-1 (CHI3L1), an enzymatically inactive N-glycosylated chitinase that is overexpressed in the colon during inflammation. Interestingly, AIEC strains harbor ChiA variants with 5 specific polymorphisms that confer increased binding to CHI3L1 compared to those in commensal K-12 strains,Citation41 supporting the role of this adhesin in promoting the virulence of CD-associated pathobionts.

Along with their main function of mediating swimming motility, flagella also play a key role in facilitating cell adhesion and invasion by AIEC.Citation28,Citation44 Compared to commensal E. coli, AIEC strains are hyperflagellated, which is congruent with their superior ability to penetrate the intestinal crypts.Citation28 Interestingly, deleting the flagellin-encoding gene in AIEC results in low piliation due to the inversion of a genetic switch in the fim promoter, suggesting a regulatory crosstalk between type I pili and flagella.Citation45 Given that both surface organelles promote cell adhesion, it is possible that the elements controlling their expression were wired during reductive evolution to generate a regulatory node that can respond to common host cues. Together, these data highlight the ability of AIEC strains to deploy multiple adhesins that can facilitate host cell contact. Nonetheless, such contact is contingent on the ability of these bacteria to overcome the mucus layer covering the epithelial cells. While flagella propel AIEC across the mucin, the ability of these bacteria to secrete mucinolytic enzymes, like Vat, further promotes mucosal invasion. This protease belongs to the SPATE family (Serine Protease Autotransporters of the Enterobacteriaceae) and is significantly overrepresented in CD-associated E. coli relative to commensals.Citation46 Moreover, Vat is upregulated in vitro under conditions that mimic the ileum, highlighting the influence of the anatomical microenvironments on virulence gene expression.Citation46

In addition to cell adhesion, AIEC strains are known for invading the host cells despite lacking type III secretion systems, which are commonly used by enteric pathogens to trigger their uptake.Citation20,Citation21 This can be explained by the invasins harbored within AIEC strains. This includes IbeA, an outer membrane invasin that interacts with host receptors such as vimentin. IbeA was found to be crucial for the ability of AIEC to invade the host cells and colonize the murine gut.Citation47 Interestingly, these findings were confirmed by a recent analysis of the in vivo transcriptome of AIEC, which revealed that ibeA is upregulated during the early phase of host infection.Citation48 Taken together, these data suggest that the fitness of AIEC in the CD environment is highly reliant on their access to the epithelial cells.

The predilection of AIEC to invade the gut epithelia could be explained by several factors. First, invading the epithelial cells and establishing an intracellular niche is an efficient mechanism to evade the phagocytic immune cells in the gut. This is further facilitated by the defects in the microbial sensing mechanisms, which are common among CD patients.Citation49 Second, the epithelial microenvironment provides AIEC with access to mucosal metabolites that are beyond the reach of the autochthonous microbes residing in the gut lumen. For example, AIEC can utilize some of the host-derived amino acids only when they are in close association with the epithelial cells.Citation50 Thus, the egress of the AIEC population, or a subpopulation, from the gut lumen toward a microenvironment with less residing microbes allows them to thrive in a less competitive niche. Indeed, the intracellular lifestyle of AIEC is not limited to epithelial cells. Since their discovery, AIEC strains were recognized for their ability to replicate within macrophages at comparable levels to that of known intracellular pathogens like Shigella and Salmonella.Citation51 However, these pathogens hijack the host defense responses using their type III secretion systems, which are absent in AIEC. This suggests that AIEC strains employ unique mechanisms to establish a replicative niche inside the host cells.Citation21,Citation22 Data from different groups suggest that the stress response is key to the survival of AIEC in macrophages. For example, the stress protein, HtrA, is required by AIEC to survive in the hostile phagolysosomal environment, potentially through its housekeeping role in degrading misfolded proteins. Interestingly, htrA was upregulated by AIEC, but not commensal E. coli, inside macrophages, further confirming the improved ability of CD-associated pathobionts to persist in hostile host environments.Citation52 Furthermore, recent work revealed that some AIEC strains are able to assemble intracellular biofilms, which fosters their ability to replicate inside macrophages.Citation53 The authors used transcriptomics to track gene expression by the intracellular bacteria. Expectedly, the genes involved in the response to oxidative stress and acidic pH are upregulated by the intracellular bacterial population. This includes the SOS response that is triggered by the genotoxic damage encountered by AIEC inside the vacuole. Additionally, the survival of AIEC inside the phagolysosomal compartment required an intact stringent response, with the latter mediating the emergence of a quiescent subpopulation that is tolerant to antibiotics.Citation54 Similarly, persister cell formation by the enteric pathogen, Salmonella enterica,Citation55 was previously reported suggesting that this is a common trait among intracellular pathogens. However, members of the AIEC pathovar display significant variability in cell invasion and intracellular replication.Citation22 Thus, a universal role for stringent and SOS responses among members of the AIEC group remains to be demonstrated.

Immune evasion by AIEC

The gut environment is equipped with several defensive mechanisms that impede the luminal bacteria from breaching the mucosal barrier. In addition to the mucus layer that physically separates most members of the gut microbiome from the underlying epithelial cells, phagocytes such as macrophages and conventional dendritic cells are continuously surveilling the mucosa for invading pathogens.Citation56 Therefore, it is not surprising that some of the risk alleles associated with CD encompass key components of the immune system that are required for sensing and eliminating bacteria.Citation49 This includes autophagy, an important housekeeping process that is required for recycling different cellular organelles and eliminating intracellular pathogens.Citation57 Missense variants of key autophagy genes, ATG16L1 and IRGM, are associated with an increased risk of CD.Citation6,Citation58 Additionally, the EIF2AK4-EIF2-ATF4 pathway, which regulates autophagy, is suppressed during CD-associated inflammation, causing impaired eradication of intracellular bacteriaCitation59 . In line with the role of autophagy in innate immunity, many pathogens have acquired the ability to hijack this pathway during their evolution.Citation60 In this regard, AIEC strains suppress autophagy by increasing the expression of the host regulatory microRNA molecules, Mir30c and Mir130a. Upon overexpression, Mir30c and Mir130a downregulate ATG16L1 and ATG5, which are essential components of the autophagy response. During the early stages of autophagy, ATG16L1 will form a complex with ATG5 and ATG12, mediating membrane expansion to form the autophagosome.Citation61 Clinical data support the role of Mir30c and Mir130a in modulating autophagy during inflammation as both microRNA molecules were overexpressed in the ileal biopsies obtained from CD patients. While the authors did not demonstrate that these tissues were infected with AIEC, the data highlight the potential of CD-associated bacteria to curtail autophagy, promoting their persistence in the intracellular environment.Citation62

In addition to autophagy, the secretion of antimicrobial peptides by Paneth cells in the gut is one of the pillars of innate mucosal immunity.Citation63 By disrupting the integrity of bacterial membranes, antimicrobial peptides limit the numbers of microbes growing at the intestinal epithelial surface. Some of the risk alleles associated with CD were found to affect the functions of Paneth cells. For example, murine models lacking NOD2, a common target for polymorphism in CD patients, display lower levels of α -defensin secretion compared to wild-type mice.Citation64 This reduction is dependent on the microbiome as Nod2 −/− mice co-housed with wild-type littermates displayed similar levels of α -defensin expression, highlighting the role of the microbiome in disease development.Citation65 Furthermore, the clinical data suggest that the secretion of antimicrobial peptides is altered during CD. When the expression of antimicrobial peptides was compared between CD patients and healthy controls, lower levels of α -defensins were detected in the biopsies obtained from the patients presenting ileal disease. Interestingly, the low expression of α -defensins correlated with NOD2/CARD15 mutations, supporting the role of NOD2 in antimicrobial peptide production.Citation66 Similar to ⍺-defensins, β-defensins like hBD-1,2,3 are differentially expressed in the guts of CD patients relative to the healthy population. While lower levels of hBD1 were detected in CD biopsies relative to healthy tissues, the opposite trend was observed for hBD 2 and 3.Citation67 These data suggest that Paneth cell functions are modulated rather than diminished during CD. It is possible that the changes in antimicrobial peptide composition during CD contribute to the dysbiosis that characterizes the disease. In this regard, the predominant host defense peptides in the CD environment will select for the resistant bacterial strains, while driving the demise of those that are susceptible. In line with this hypothesis, AIEC strains are highly resistant to α- and β-defensins.Citation68 This is dependent on the presence of outer membrane protease, OmpT, and other proteins that belong to Mig-14 family, known to mediate resistance to antimicrobial peptidesCitation68,Citation69 . Interestingly, the resistance of AIEC to host defense peptides is required to exacerbate the inflammation caused by acute infectious gastroenteritis, a known risk factor for CD.Citation70 Using a polymicrobial infection model, Small et al showed that AIEC can worsen the inflammation triggered by enteric pathogens, like Salmonella typhimurium and Citrobacter rodentium. Unlike the wild-type AIEC, an isogenic mutant susceptible to host defense peptides showed less morbidity when the animals were co-infected with S. typhimurium,Citation71 confirming that antimicrobial peptide resistance is key for the ability of CD-associated pathobionts to aggravate inflammation.

Metabolic adaptation of AIEC

The ability of pathogens to utilize the metabolites available in the gut during inflammation is key to their expansion at the expense of the healthy microbiome.Citation72,Citation73 Similar to several enteric pathogens, the consumption of inflammation-associated metabolites by AIEC contributes significantly to their success in colonizing the gut (). During inflammation, there is an increased production of nitric oxide, which can react with superoxide to generate peroxynitrite. Due to its instability, peroxynitrite can decompose into nitrate, or reacts with organic sulfides and tertiary amines, releasing S-Oxides and N-Oxides into the gut.Citation74 Unlike strict anaerobes, facultative anaerobes, including members of family Enterobacteriaceae, can use nitrates, S-Oxides and N-Oxides as terminal electron acceptors during respiration.Citation75 This provides Enterobacteriaceae, like E. coli, a metabolic advantage in the inflamed gut. Indeed, nitrate utilization fuels the expansion of AIEC during inflammation.Citation76 An AIEC mutant deficient in utilizing nitrate as a terminal electron acceptor displayed lower fitness in the host environment compared to wild-type isogenic strain. Similarly, the consumption of oxidized sugars, like glucarate and galactarate, is instrumental for AIEC to thrive in the gut during the inflammation that ensues from antibiotic administration, a common treatment to manage CD flares.Citation76 These results were confirmed using a high-resolution in vivo genetic screen that described the fitness landscape of AIEC in the gut.Citation48 Using a combined genomics approach, genes that are essential for host colonization and are expressed by AIEC early during infection were mapped. Indeed, this analysis confirmed that metabolic adaptation is central to the ability of AIEC to thrive in the gut.Citation48

Figure 1. Examples of the key metabolites utilized by AIEC in the gut environment.

Figure 1. Examples of the key metabolites utilized by AIEC in the gut environment.

To investigate the metabolic response of AIEC to inflammation, Kitamoto et al mapped the bacterial transcriptome during severe colitis.Citation77 Many pathways involved in amino acid metabolism were specifically upregulated during inflammation compared to normal intestinal conditions. This includes the pathways for utilizing L-serine, which was found to be critical for the ability of AIEC to expand in the inflamed intestine and to exacerbate colitis.Citation77 This fitness advantage is contingent on the availability of L-serine in the gut. Accordingly, the deprivation of dietary L-serine tied the fitness of AIEC to the presence of mucolytic symbionts that can degrade the host mucins to liberate this amino acid.Citation50 Importantly, these studies highlight how diet can modulate the behavior of AIEC in vivo. Similarly, a Western diet that is high in fat and sugar promotes the expansion of AIEC in the gut.Citation78,Citation79 Interestingly, the low fiber content of Western diet was shown to be the main driver of AIEC expansion compared to high fat and sugar content, uncovering the potential of dietary intervention to combat the CD-associated pathogens.Citation79

Other gut metabolites consumed by AIEC strains include fucose and its fermentation product, 1,2-propanediol.Citation80 Using the propanediol dehydratase, PduC, E. coli can convert 1,2-propanediol to propionate, which can be further metabolized to pyruvate to be oxidized in the tricarboxylic acid cycle. Comparative genomics revealed that the pdu operon is more enriched in the genomes of AIEC relative to commensal E. coli.Citation81 In agreement with its role in promoting AIEC expansion in the gut, PduC is upregulated in vitro in the presence of bile salts, commonly present in the intestine.Citation80 Similarly, bile salts induced the eut operon, required for AIEC to utilize ethanolamine as a nitrogen source.Citation80 Ethanolamine is released from phosphatidylethanolamine, which is highly abundant in the cell membranes of the host and its microbiome. Using clinical isolates, Zhang et al showed that AIEC strains can utilize ethanolamine as both carbon and nitrogen sources. Of note, this was enhanced by the presence of other inflammation-associated metabolites.Citation82 Additionally, the authors investigated how inflammation-associated metabolites can influence global gene expression in AIEC, showing that some metabolites can upregulate virulence genes. However, it is unclear if this in vitro analysis approximates the transcriptomes of bacteria in vivo, given the complexity of the metabolic landscape in the gut. Nevertheless, other studies suggest that ethanolamine utilization supports AIEC blooms in the gut. For example, lipid precursors of ethanolamine, N-acylethanolamines, are elevated in the gut of IBD patients and preclinical models of colitis, highlighting the importance of this metabolite in fueling the expansion of CD-associated bacteria.Citation83 These results were further confirmed by a recent study that mapped the in vivo transcriptome of AIEC, showing that several ethanolamine utilization genes are upregulated by these bacteria in the intestine.Citation48 While these data corroborate the importance of ethanolamine to AIEC fitness, some strains have lost the ability to utilize this metabolite due to deletions in some eut genes,Citation82 highlighting the distinct adaptive routes of the different AIEC strains in the gut.

The metabolic plasticity of AIEC includes the consumption of short chain fatty acids (SCFAs),Citation28 an important class of metabolites that is cardinal to the maintenance of intestinal homeostasis.Citation84,Citation85 In addition to being the preferred energy source for colonocytes,Citation86 SCFAs lower the inflammatory tone of the gut.Citation84 For example, butyrate, and less efficiently propionate, can inhibit histone deacetylases promoting the differentiation of Foxp3+ T regulatory cells, known to suppress inflammation.Citation87,Citation88 Additionally, the binding of SCFAs to G-protein coupled receptors (GPCR) 43 and 109A was shown to modulate T cell differentiation.Citation89,Citation90 In this regard, propionate enhanced the expression of Foxp3 and IL-10 in murine T regulatory cells only when GPCR-43 was present.Citation90 Similarly, the binding of butyrate to GPCR-109A is important to induce the differentiation of IL-10 producing T regulatory cells.Citation89 Therefore, it is not surprising that low endogenous concentrations of SCFAs have been associated with gut inflammation.Citation91 In line with their pro-inflammatory behavior, AIEC are more adept than commensal E. coli in consuming SCFAs as carbon source,Citation28 potentially leading to a decreased abundance of these anti-inflammatory molecules in vivo. This can potentially explain why CD patients have less intestinal SCFAs than healthy individuals.Citation91–93 While this can be driven by the reduced abundance of fiber-fermenting bacteria in the gut,Citation91 it remains possible that AIEC can further deplete SCFAs through catabolism. Moreover, the role of SCFAs in promoting AIEC fitness extends beyond providing these bacteria with energy. Upon exposure to butyrate or propionate, AIEC strains display increased adhesion and invasion of the epithelial cells.Citation94 This is due to the upregulation of flagellar gene expression, which is known to potentiate the invasiveness of AIEC.Citation28 Similarly, propionate was shown to promote the virulence and persistence of AIEC in the gut.Citation95,Citation96 Interestingly, propionate did not increase the adhesion and invasion of epithelial cells by commensal E. coli, suggesting that this adaptation is unique to AIEC.Citation96 However, the modulation of AIEC virulence by SCFAs was shown to be largely dependent on the gut biogeography.Citation97 For example, flagellar motility is increased under pH and SCFA concentrations mimicking that of the ileum, while the inverse is observed with colon mimicry.Citation97 Therefore, more work is required to uncover the influence of the different gut microenvironments on the pathoadaptation of AIEC. While the genetic determinants for host colonization by AIEC were recently identified,Citation48 approaches with higher resolution are required to investigate how bacterial fitness varies across the gut landscape.

In addition to the competition for carbon and nitrogen sources, the battle for metals is equally important for the success of pathogens to colonize the gut.Citation98 Since many metabolic enzymes require metals, like iron, as co-factors, their acquisition is essential for bacteria. Comparative genomics revealed that the genomes of AIEC strains are enriched in iron acquisition genes relative to commensals.Citation81 This includes heme utilization genes and siderophores. Upon their secretion to the extracellular environment, siderophores bind iron with high affinity.Citation98 Using dedicated transporters, bacteria can uptake the iron-bound siderophores and shuttle them to the cytoplasm, where iron is liberated. Transcriptomics revealed the upregulation of several siderophore synthesis and transport genes by AIEC in the intestinal tissuesCitation48 and inside macrophages.Citation53 This includes Yersiniabactin, a stealth siderophore that can evade sequestration by Lipocalin-2, a host defense protein known to chelate other siderophores.Citation98,Citation99 Interestingly, Yersiniabactin was shown to be required for the induction of fibrosis by AIEC in Il10−/− mice, suggesting that this siderophore has immunomodulatory roles.Citation100 This is in line with other studies showing that siderophores can interfere with immune functions. For example, enterobactin can inhibit myeloperoxidase, the enzyme responsible for generating the potent anti-microbial agent, hypochlorous acid.Citation101 In addition to Yersiniabactin, AIEC strains carry other stealth siderophores, like Salmochelin. While this siderophore is dispensable for AIEC fitness in the gut during homeostasis, it is key for the expansion of these bacteria during psychological stress,Citation102 a disease modifier associated with CD flares.Citation103 This conditional essentiality of siderophores sheds light on their niche-specific roles, providing an explanation for their apparent functional redundancy. Nevertheless, these data highlight the potential of siderophores as targets for novel therapeutics. In accord with this, host immunization with siderophore-based vaccines was shown to elicit a strong mucosal immune response that can quell AIEC expansion and decrease gut inflammation.Citation104 Taken together, these results underpin the potential of targeting iron acquisition systems as a strategy to complement the host nutritional immunity against CD-associated pathogens.

The role of biofilms in promoting the persistence of AIEC

By embedding themselves in a self-produced extracellular matrix, bacteria can form biofilms on abiotic and biotic surfaces.Citation105–107 The biofilm matrix constitutes proteins, exopolysaccharides, and DNA, which collectively protect the enclosed bacteria from external threats, including the immune cells.Citation108 Indeed, the comparison of the spatial organization of bacteria during health and disease suggests that biofilm formation contributes to disease development. Using microscopy, Swidsinski et al showed that bacterial biofilms are more abundant in the mucosa of CD patients relative to healthy individuals by approximately two orders of magnitude.Citation109 These dense biofilms were detected in the guts of 95% of the IBD patients, while only 35% of the healthy individuals had mucosal biofilms. Whether these biofilms are populated by healthy members of the microbiome or pathobionts remains undetermined. However, this triggered interest in exploring the ability of CD-associated pathogens, like AIEC, to form biofilms. In this regard, the archetypal AIEC strain LF82 was shown to form biofilms on the surface of intestinal epithelial cells, a process that requires the σE stress response. A strain overexpressing anti- σE factors formed less biofilms on epithelial cells. However, this strain displays impaired adhesion to host cells, making the direct contribution of σE to biofilm formation challenging to quantify.Citation110 A recent study has identified biofilm formation as one of the key elements of AIEC fitness in the gut.Citation48 This process requires a functional type IV secretion system (T4SS), which is employed by AIEC to form microcolonies on the epithelial surface. T4SS in bacteria comprises inner and outer membrane complexes that elaborate pili, which function in bringing the mating bacteria to proximity to allow DNA transfer.Citation111–113 Although traditionally known for its role in conjugation, T4SS facilitates biofilm formation by E. coli both in vitroCitation114 and in vivo.Citation48 The enrichment of T4SS-encoding genes in the microbial metagenomes of CD patients suggests a role in disease development, potentially through promoting the ability of pathobionts to form biofilms and persist in the gut. CD is characterized by recurrent episodes of inflammation that are associated with bacterial expansion. One possible explanation for these flares is the presence of persistent bacterial communities that are shielded by their biofilms from the immune system. In the presence of specific environmental cues, these bacteria can then disseminate from their biofilms, launching a pro-inflammatory response in the gut, driving CD flares. In accord with this, biofilm production by AIEC was shown to be important for triggering inflammation in colitogenic mouse models.Citation115 In addition to forming biofilms on mucosal surfaces, some AIEC strains like LF82 are able to form intracellular biofilm-like structures.Citation53 During late stages of infection, the intracellular AIEC population induces a suite of genes required for biofilm formation inside the phagosomal compartment of macrophages. These intracellular biofilms were found to be pivotal for the survival of AIEC inside macrophages, which is a key trait that discriminates these bacteria from commensal E. coli.Citation22,Citation51

Conclusions and future directions

Given the multifactorial nature of CD, it is challenging to fulfill Koch’s postulates when studying the contribution of microbes to its etiology. The perpetual complexity of the gut ecosystem, together with the influence of host genetics and environmental factors on the microbiome, limit our ability to decipher the role of particular organisms in driving inflammation. Traditionally, microbes have been viewed as a dichotomy of pathogens and commensals. Nevertheless, it is clear now that virulence can spawn within susceptible hosts as an emergent property.Citation33 The AIEC group has received significant attention due to the high association of these strains with the CD environmentCitation18,Citation19 and their ability to trigger inflammation in preclinical models.Citation30,Citation32 However, the lack of a clear evolutionary origin for this group suggests that the adherent-invasive phenotype might be an emergent property in the susceptible hosts.Citation116 The isolation of AIEC from healthy individuals, while less frequently compared to CD patients, suggests that their pathogenesis is contextual. In line with this hypothesis, experimental evolution showed that the host environment selects for several pathoadaptive traits that promote the ability of AIEC to adhere to and invade the host cells.Citation28 Similarly, the juxtaposition of host susceptibility and pathobiont evolution can lead to the emergence of detrimental microbial traits among commensals.Citation117 Indeed, it is important to study the biochemical mechanisms underlying the virulence of AIEC. Nevertheless, it is equally important to unearth the triggers of emergent virulence among gut bacteria to develop a better understanding of CD etiology. This is critical to guide the drug screening pipelines aiming at developing selective therapies that can eradicate CD-associated pathobionts while maintaining the healthy microbiome.

Disclosure statement

No potential conflicts of interest were disclosed.

Data availability statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Correction Statement

This article has been corrected with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

W.E. is Canada Research Chair in Microbiome Research (Grant I.D. RES0056038RS). The work in Elhenawy laboratory is funded by Li Ka Shing Institute of Virology (Grant I.D. RES0057474) and Major Innovation Fund - One Health Consortium, Alberta (Grant I.D. RES0059404).

References

  • O’Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Rep. 2006;7:688–14. doi:10.1038/sj.embor.7400731.
  • Kaplan GG, Bernstein CN, Coward S, Bitton A, Murthy SK, Nguyen GC, Lee K, Cooke-Lauder J, Benchimol EI. The impact of inflammatory bowel disease in Canada 2018: epidemiology. J Canadian Ass Gastro. 2019;2:S6–16. doi:10.1093/jcag/gwy054.
  • Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46–54. e42;quiz e30. doi:10.1053/j.gastro.2011.10.001.
  • Feuerstein JD, Cheifetz AS. Crohn disease: epidemiology, diagnosis, and management. Mayo Clinic Proceedings. 2017;92:1088–1103. doi:10.1016/j.mayocp.2017.04.010.
  • Franke A, Dpb M, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, Lees CW, Balschun T, Lee J, Roberts R, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118–1125. doi:10.1038/ng.717.
  • Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, Ripke S, Lee JC, Jostins L, Shah T, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–986. doi:10.1038/ng.3359.
  • Liu T-C, Stappenbeck TS. Genetics and pathogenesis of inflammatory bowel disease. Annu Rev Pathol Mech Dis. 2016;11:127–148. doi:10.1146/annurev-pathol-012615-044152.
  • Halfvarson J, Bodin L, Tysk C, Lindberg E, Järnerot G. Inflammatory bowel disease in a Swedish twin cohort: a long-term follow-up of concordance and clinical characteristics. Gastroenterology. 2003;124:1767–1773. doi:10.1016/S0016-5085(03)00385-8.
  • Feller M, Huwiler K, Stephan R, Altpeter E, Shang A, Furrer H, Pfyffer GE, Jemmi T, Baumgartner A, Egger M. Mycobacterium avium subspecies paratuberculosis and Crohn’s disease: a systematic review and meta-analysis. Lancet Infect Dis. 2007;7:607–613. doi:10.1016/S1473-3099(07)70211-6.
  • Behr MA, Kapur V. The evidence for Mycobacterium paratuberculosis in Crohn’s disease: current Opinion in Gastroenterology. Curr Opin Gastroenterol. 2008;24:17–21. doi:10.1097/MOG.0b013e3282f1dcc4.
  • Agrawal G, Aitken J, Hamblin H, Collins M, Borody TJ. Putting Crohn’s on the MAP: five common questions on the contribution of Mycobacterium avium subspecies paratuberculosis to the pathophysiology of Crohn’s disease. Dig Dis Sci. 2021;66:348–358. doi:10.1007/s10620-020-06653-0.
  • Bull TJ, McMinn EJ, Sidi-Boumedine K, Skull A, Durkin D, Neild P, Rhodes G, Pickup R, Detection H-TJ. Verification of Mycobacterium avium subsp. J Clin Microbiol. 2003;41:2915–2923.
  • Jones PH, Farver TB, Beaman B, Çetinkaya B, Morgan KL. Crohn’s disease in people exposed to clinical cases of bovine paratuberculosis. Epidemiol Infect. 2006;134:49–56. doi:10.1017/S0950268805004681.
  • Selby W, Pavli P, Crotty B, Florin T, Radford-Smith G, Gibson P, Mitchell B, Connell W, Read R, Merrett M, et al. Two-Year combination antibiotic therapy with Clarithromycin, Rifabutin, and Clofazimine for Crohn’s Disease. Gastroenterology. 2007;132:2313–2319. doi:10.1053/j.gastro.2007.03.031.
  • Liu Y, Van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel J-F. Immunocytochemical evidence of Listeria, Escherichia coil, and Streptococcus antigens in Crohn’s disease. Gastroenterology. 1995;108:1396–1404. doi:10.1016/0016-5085(95)90687-8.
  • Magin WS, Van Kruiningen HJ, Colombel J-F. Immunohistochemical search for viral and bacterial antigens in Crohn’s disease. J Crohn’s Colitis. 2013;7:161–166. doi:10.1016/j.crohns.2012.03.021.
  • Darfeuille-Michaud A, Neut C, Barnich N, Lederman E, Di Martino P, Desreumaux P, Gambiez L, Joly B, Cortot A, Colombel JF. Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn’s disease. Gastroenterology. 1998;115:1405–1413. doi:10.1016/S0016-5085(98)70019-8.
  • Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser A-L, Barnich N, Bringer M-A, Swidsinski A, Beaugerie L, Colombel J-F. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004;127:412–421. doi:10.1053/j.gastro.2004.04.061.
  • Nadalian B, Yadegar A, Houri H, Olfatifar M, Shahrokh S, Asadzadeh Aghdaei H, Suzuki H, Zali MR. Prevalence of the pathobiont adherent-invasive Escherichia coli and inflammatory bowel disease: a systematic review and meta-analysis. J Gastroenterol Hepatol. 2021;36:852–863. doi:10.1111/jgh.15260.
  • Desilets M, Deng X, Deng X, Rao C, Ensminger AW, Krause DO, Sherman PM, Gray-Owen SD. Genome-based definition of an inflammatory bowel disease-associated adherent-Invasive Escherichia coli Pathovar. Inflamm Bowel Dis. 2016;22:1–12. doi:10.1097/MIB.0000000000000574.
  • Nash JH, Villegas A, Kropinski AM, Aguilar-Valenzuela R, Konczy P, Mascarenhas M, Ziebell K, Torres AG, Karmali MA, Coombes BK. Genome sequence of adherent-invasive Escherichia coli and comparative genomic analysis with other E. coli pathotypes. BMC Genomics. 2010;11:667. doi:10.1186/1471-2164-11-667.
  • O’Brien CL, Bringer M-A, Holt KE, Gordon DM, Dubois AL, Barnich N, Darfeuille-Michaud A, Pavli P. Comparative genomics of Crohn’s disease-associated adherent-invasive Escherichia coli. Gut. 2017;66:1382–1389. doi:10.1136/gutjnl-2015-311059.
  • Camprubí-Font C, Martinez-Medina M. Why the discovery of adherent-invasive Escherichia coli molecular markers is so challenging? WJBC. 2020;11:1–13. doi:10.4331/wjbc.v11.i1.1.
  • Martinez-Medina M, Aldeguer X, Lopez-Siles M, González-Huix F, López-Oliu C, Dahbi G, Blanco JE, Blanco J, Garcia-Gil LJ, Darfeuille-Michaud A. Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn’s disease. Inflamm Bowel Dis. 2009;15:872–882. doi:10.1002/ibd.20860.
  • Costa RFA, Ferrari MLA, Bringer M-A, Darfeuille-Michaud A, Martins FS, Barnich N. Characterization of mucosa-associated Escherichia coli strains isolated from Crohn’s disease patients in Brazil. BMC Microbiol. 2020;20:178. doi:10.1186/s12866-020-01856-x.
  • Kotlowski R, Bernstein CN, Sepehri S, Krause DO. High prevalence of Escherichia coli belonging to the B2+D phylogenetic group in inflammatory bowel disease. Gut. 2007;56:669–675. doi:10.1136/gut.2006.099796.
  • Conte MP, Longhi C, Marazzato M, Conte AL, Aleandri M, Lepanto MS, Zagaglia C, Nicoletti M, Aloi M, Totino V, et al. Adherent-invasive Escherichia coli (AIEC) in pediatric Crohn’s disease patients: phenotypic and genetic pathogenic features. BMC Res Notes. 2014;7:748. doi:10.1186/1756-0500-7-748.
  • Elhenawy W, Tsai CN, Coombes BK. Host-Specific adaptive diversification of Crohn’s Disease-Associated adherent-Invasive Escherichia coli. Cell Host Microbe. 2019;25(301–312.e5):301–312.e5. doi:10.1016/j.chom.2018.12.010.
  • Carvalho FA, Barnich N, Sauvanet P, Darcha C, Gelot A, Darfeuille-Michaud A. Crohn’s disease-associated Escherichia coli LF82 aggravates colitis in injured mouse colon via signaling by flagellin. Inflamm Bowel Dis. 2008;14:1051–1060. doi:10.1002/ibd.20423.
  • Small CLN, Reid-Yu SA, McPhee JB, Coombes BK, Small C-LN, Reid-Yu SA, McPhee JB, Coombes BK. Persistent infection with Crohn’s disease-associated adherent-invasive Escherichia coli leads to chronic inflammation and intestinal fibrosis. Nat Commun. 2013;4:1957. 10.1038/ncomms2957.
  • Chassaing B, Koren O, Carvalho FA, Ley RE, Gewirtz AT. AIEC pathobiont instigates chronic colitis in susceptible hosts by altering microbiota composition. Gut. 2014;63:1069–1080. doi:10.1136/gutjnl-2013-304909.
  • Schmitz JM, Tonkonogy SL, Dogan B, Leblond A, Whitehead KJ, Kim SC, Simpson KW, Sartor RB. Murine adherent and invasive E. coli induces chronic inflammation and immune responses in the small and large intestines of monoassociated IL-10-/- Mice independent of long polar fimbriae adhesin. Inflamm Bowel Dis. 2019;25(5): 875–885.
  • Hajishengallis G, Lamont RJ. Dancing with the stars: how choreographed bacterial interactions dictate nososymbiocity and give rise to keystone pathogens, accessory pathogens, and pathobionts. Trends Microbiol. 2016;24:477–489. doi:10.1016/j.tim.2016.02.010.
  • Barnich N, Carvalho FA, Glasser A-L, Darcha C, Jantscheff P, Allez M, Peeters H, Bommelaer G, Desreumaux P, Colombel J-F, et al. CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J Clin Invest. 2007;117:1566–1574. doi:10.1172/JCI30504.
  • Åv K, Alkaissi LY, Holm EB, Heil SDS, Chassaing B, Darfeuille-Michaud A, McKay DM, Söderholm JD. Enhanced E. coli LF82 translocation through the Follicle-associated Epithelium in Crohn’s disease is dependent on long polar fimbriae and CEACAM6 expression, and increases paracellular permeability. J Crohn’s Colitis. 2020;14:216–229. doi:10.1093/ecco-jcc/jjz144.
  • Dreux N, Denizot J, Martinez-Medina M, Mellmann A, Billig M, Kisiela D, Chattopadhyay S, Sokurenko E, Neut C, Gower-Rousseau C, et al. Point mutations in FimH adhesin of Crohn’s disease-associated adherent-invasive Escherichia coli enhance intestinal inflammatory response. PLoS Pathog. 2013;9:e1003141. doi:10.1371/journal.ppat.1003141.
  • Chevalier G, Laveissière A, Desachy G, Barnich N, Sivignon A, Maresca M, Nicoletti C, Di Pasquale E, Martinez-Medina M, Simpson KW, et al. Blockage of bacterial FimH prevents mucosal inflammation associated with Crohn’s disease. Microbiome. 2021;9:176. doi:10.1186/s40168-021-01135-5.
  • Sivignon A, Yan X, Alvarez Dorta D, Bonnet R, Bouckaert J, Fleury E, Bernard J, Gouin SG, Darfeuille-Michaud A, Barnich N. Development of Heptylmannoside-Based Glycoconjugate Antiadhesive compounds against adherent-Invasive Escherichia coli bacteria associated with Crohn’s Disease. mBio. 2015;6:e01298–15. doi:10.1128/mBio.01298-15.
  • Rolhion N, Carvalho FA, OmpC D-MA. and the sigma(E) regulatory pathway are involved in adhesion and invasion of the Crohn’s disease-associated Escherichia coli strain LF82. Mol Microbiol. 2007;63:1684–1700. doi:10.1111/j.1365-2958.2007.05638.x.
  • Fdm VS, Oldenburg B, Hart AR, Siersema PD, Lindgren S, Grip O, Teucher B, Kaaks R, Bergmann MM, Boeing H, et al. Serological markers predict inflammatory bowel disease years before the diagnosis. Gut. 2013;62:683–688. doi:10.1136/gutjnl-2012-302717.
  • Low D, Tran HT, Lee I-A, Dreux N, Kamba A, Reinecker H-C, Darfeuille-Michaud A, Barnich N, Mizoguchi E. Chitin-binding domains of Escherichia coli ChiA mediate interactions with intestinal epithelial cells in mice with colitis. Gastroenterology. 2013;145(602–612.e9). doi:10.1053/j.gastro.2013.05.017.
  • Kawada M, Chen -C-C, Arihiro A, Nagatani K, Watanabe T, Chitinase ME. 3-like-1 enhances bacterial adhesion to colonic epithelial cells through the interaction with bacterial chitin-binding protein. Lab Invest. 2008;88:883–895. doi:10.1038/labinvest.2008.47.
  • Kirn TJ, Jude BA, Taylor RK. A colonization factor links Vibrio cholerae environmental survival and human infection. Nature. 2005;438:863–866. doi:10.1038/nature04249.
  • Sevrin G, Massier S, Chassaing B, Agus A, Delmas J, Denizot J, Billard E, Barnich N. Adaptation of adherent-invasive E. coli to gut environment: impact on flagellum expression and bacterial colonization ability. Gut Microbes. 2020;11:364–380. doi:10.1080/19490976.2017.1421886.
  • Barnich N, Boudeau J, Claret L, Darfeuille-Michaud A. Regulatory and functional co-operation of flagella and type 1 pili in adhesive and invasive abilities of AIEC strain LF82 isolated from a patient with Crohn’s disease. Mol Microbiol. 2003;48:781–794. doi:10.1046/j.1365-2958.2003.03468.x.
  • Gibold L, Garenaux E, Dalmasso G, Gallucci C, Cia D, Mottet-Auselo B, Faïs T, Darfeuille-Michaud A, Nguyen HTT, Barnich N, et al. The Vat-AIEC protease promotes crossing of the intestinal mucus layer by Crohn’s disease-associated Escherichia coli. Cell Microbiol. 2016;18:617–631. doi:10.1111/cmi.12539.
  • Cieza RJ, Hu J, Ross BN, Sbrana E, Torres AG, Payne SM. The IbeA invasin of adherent-invasive Escherichia coli mediates interaction with intestinal epithelia and macrophages. Infect Immun. 2015;83:1904–1918. doi:10.1128/IAI.03003-14.
  • Elhenawy W, Hordienko S, Gould S, Oberc AM, Tsai CN, Hubbard TP, Waldor MK, Coombes BK. High-throughput fitness screening and transcriptomics identify a role for a type IV secretion system in the pathogenesis of Crohn’s disease-associated Escherichia coli. Nat Commun. 2021;12:2032. doi:10.1038/s41467-021-22306-w.
  • Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, Brant SR, Silverberg MS, Taylor KD, Barmada MM, the NIDDK IBD Genetics Consortium, the Belgian-French IBD Consortium, the Wellcome Trust Case Control Consortium, Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, Brant SR, et al. Genome-wide association defines more than. 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40:955–962. 10.1038/ng.175.
  • Sugihara K, Kitamoto S, Saraithong P, Nagao-Kitamoto H, Hoostal M, McCarthy C, Rosevelt A, Muraleedharan CK, Gillilland MG, Imai J, et al. Mucolytic bacteria license pathobionts to acquire host-derived nutrients during dietary nutrient restriction. Cell Rep. 2022;40:111093. doi:10.1016/j.celrep.2022.111093.
  • Glasser AL, Boudeau J, Barnich N, Perruchot MH, Colombel JF, Darfeuille-Michaud A, Tuomanen EI. Adherent Invasive Escherichia coli Strains from Patients with Crohn’s Disease Survive and Replicate within Macrophages without Inducing Host Cell Death. Infect Immun. 2001;69:5529–5537. doi:10.1128/IAI.69.9.5529-5537.2001.
  • Bringer M-A, Barnich N, Glasser A-L, Bardot O, Darfeuille M-A. HtrA stress protein is involved in intramacrophagic replication of adherent and invasive Escherichia coli strain LF82 isolated from a patient with Crohn’s disease. Infect Immun. 2005;73:712–721. doi:10.1128/IAI.73.2.712-721.2005.
  • Prudent V, Demarre G, Vazeille E, Wery M, Quenech’Du N, Ravet A, Dauverd - Girault J, van Dijk E, M-A B, Descrimes M, et al. The Crohn’s disease-related bacterial strain LF82 assembles biofilm-like communities to protect itself from phagolysosomal attack. Commun Biol. 2021;4:627. doi:10.1038/s42003-021-02161-7.
  • Demarre G, Prudent V, Schenk H, Rousseau E, Bringer M-A, Barnich N, Tran Van Nhieu G, Rimsky S, De MS, Espéli O. The Crohn’s disease-associated Escherichia coli strain LF82 relies on SOS and stringent responses to survive, multiply and tolerate antibiotics within macrophages. PLoS Pathog. 2019;15:e1008123. doi:10.1371/journal.ppat.1008123.
  • Helaine S, Cheverton AM, Watson KG, Faure LM, Matthews SA, Holden DW. Internalization of Salmonella by macrophages induces formation of nonreplicating persisters. Science. 2014;343:204–208. doi:10.1126/science.1244705.
  • Perez-Lopez A, Behnsen J, Nuccio S-P RM. Mucosal immunity to pathogenic intestinal bacteria. Nat Rev Immunol. 2016;16:135–148. doi:10.1038/nri.2015.17.
  • Deretic V. Autophagy in inflammation, infection, and immunometabolism. Immunity. 2021;54:437–453. doi:10.1016/j.immuni.2021.01.018.
  • Hampe J, Franke A, Rosenstiel P, Till A, Teuber M, Huse K, Albrecht M, Mayr G, De La Vega FM, Briggs J, et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet. 2007;39:207–211. doi:10.1038/ng1954.
  • Bretin A, Carrière J, Dalmasso G, Bergougnoux A, B’chir W, Maurin A-C, Müller S, Seibold F, Barnich N, Bruhat A, et al. Activation of the EIF2AK4-EIF2A/eIF2α-ATF4 pathway triggers autophagy response to Crohn disease-associated adherent-invasive Escherichia coli infection. Autophagy. 2016;12:770–783. doi:10.1080/15548627.2016.1156823.
  • Asrat S, de Jesús DA, Hempstead AD, Ramabhadran V, Isberg RR. Bacterial Pathogen Manipulation of host membrane trafficking. Annu Rev Cell Dev Biol. 2014;30:79–109. doi:10.1146/annurev-cellbio-100913-013439.
  • Sheng Y, Song Y, Li Z, Wang Y, Lin H, Cheng H, Zhou R. RAB37 interacts directly with ATG5 and promotes autophagosome formation via regulating ATG5-12-16 complex assembly. Cell Death Differ [Internet] 2017 [cited 2022 Sep 21]; Available from]. http://www.nature.com/articles/s41418-017-0023-1
  • Nguyen HTT, Dalmasso G, Müller S, Carrière J, Seibold F, Darfeuille-Michaud A. Crohn’s disease-associated adherent invasive Escherichia coli modulate levels of microRNAs in intestinal epithelial cells to reduce autophagy. Gastroenterology. 2014;146:508–519. doi:10.1053/j.gastro.2013.10.021.
  • Bevins CL, Salzman NH. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nat Rev Micro. 2011;9:356–368. doi:10.1038/nrmicro2546.
  • Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nuñez G, Flavell RA. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307:731–734. doi:10.1126/science.1104911.
  • Shanahan MT, Carroll IM, Grossniklaus E, White A, von Furstenberg RJ, Barner R, Fodor AA, Henning SJ, Sartor RB, Gulati AS. Mouse Paneth cell antimicrobial function is independent of Nod2. Gut. 2014;63:903–910. doi:10.1136/gutjnl-2012-304190.
  • Wehkamp J, Salzman NH, Porter E, Nuding S, Weichenthal M, Petras RE, Shen B, Schaeffeler E, Schwab M, Linzmeier R, et al. Reduced Paneth cell α-defensins in ileal Crohn’s disease. Proc Natl Acad Sci USA. 2005;102:18129–18134. doi:10.1073/pnas.0505256102.
  • Wehkamp J, Harder J, Weichenthal M, Mueller O, Herrlinger KR, Fellermann K, Schroeder JM, Stange EF. Inducible and constitutive β-defensins are differentially expressed in Crohn’s Disease and Ulcerative Colitis: inflammatory bowel diseases. Inflammatory Bowel Diseases. 2003;9(4):215–223. doi:10.1097/00054725-200307000-00001.
  • McPhee JB, Small CL, Reid-Yu SA, Brannon JR, Le Moual H, Coombes BK, McPhee JB, Small CL, Reid-Yu SA, Brannon JR, Le Moual H, Coombes BK. Host defense peptide resistance contributes to colonization and maximal intestinal pathology by Crohn’s Disease-associated adherent-Invasive Escherichia coli. Infect Immun. 2014;82:3383–3393. 10.1128/IAI.01888-14.
  • Brodsky IE, Ernst RK, Miller SI, Falkow S. mig - 14 is a Salmonella gene that plays a role in bacterial resistance to antimicrobial peptides. J Bacteriol. 2002;184:3203–3213. doi:10.1128/JB.184.12.3203-3213.2002.
  • Porter CK, Tribble DR, Aliaga PA, Halvorson HA, Riddle MS. Infectious gastroenteritis and risk of developing inflammatory bowel disease. Gastroenterology. 2008;135:781–786. doi:10.1053/j.gastro.2008.05.081.
  • Small CL, Xing L, McPhee JB, Law HT, Coombes BK, Small CL, Xing L, McPhee JB, Law HT, Coombes BK. Acute infectious gastroenteritis potentiates a Crohn’s disease pathobiont to fuel ongoing inflammation in the Post-Infectious period. PLoS Pathog. 2016;12:e1005907. 10.1371/journal.ppat.1005907.
  • Bäumler AJ, Sperandio V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature. 2016;535:85–93. doi:10.1038/nature18849.
  • Miller BM, Bäumler AJ. The habitat filters of microbiota-nourishing immunity. Annu Rev Immunol. 2021;39:1–18. doi:10.1146/annurev-immunol-101819-024945.
  • Szabó C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov. 2007;6:662–680. doi:10.1038/nrd2222.
  • Winter SE, Winter MG, Xavier MN, Thiennimitr P, Poon V, Keestra AM, Laughlin RC, Gomez G, Wu J, Lawhon SD, et al. Host-Derived nitrate boosts growth of E. coli in the Inflamed Gut. Science. 2013;339:708–711. doi:10.1126/science.1232467.
  • Oberc AM, Fiebig-Comyn AA, Tsai CN, Elhenawy W, Coombes BK. Antibiotics potentiate adherent-Invasive E. coli Infection and expansion. Inflamm Bowel Diseases. 2019;25(4):711–721. doi:10.1093/ibd/izy361.
  • Kitamoto S, Alteri CJ, Rodrigues M, Nagao-Kitamoto H, Sugihara K, Himpsl SD, Bazzi M, Miyoshi M, Nishioka T, Hayashi A, et al. Dietary l-serine confers a competitive fitness advantage to Enterobacteriaceae in the inflamed gut. Nat Microbiol. 2020;5:116–125. doi:10.1038/s41564-019-0591-6.
  • Agus A, Denizot J, Thévenot J, Martinez-Medina M, Massier S, Sauvanet P, Bernalier-Donadille A, Denis S, Hofman P, Bonnet R, et al. Western diet induces a shift in microbiota composition enhancing susceptibility to Adherent-Invasive E. Sci Rep. 2016;6:19032.
  • Lau TC, Fiebig-Comyn AA, Shaler CR, McPhee JB, Coombes BK, Schertzer JD. Low dietary fiber promotes enteric expansion of a Crohn’s disease-associated pathobiont independent of obesity. American J Phy Endocrinology and Metabolism. 2021 ajpendo.00134.2021;321(3):E338–E350. doi:10.1152/ajpendo.00134.2021.
  • Delmas J, Gibold L, Faïs T, Batista S, Leremboure M, Sinel C, Vazeille E, Cattoir V, Buisson A, Barnich N, et al. Metabolic adaptation of adherent-invasive Escherichia coli to exposure to bile salts. Sci Rep. 2019;9:2175. doi:10.1038/s41598-019-38628-1.
  • Dogan B, Suzuki H, Herlekar D, Sartor RB, Campbell BJ, Roberts CL, Stewart K, Scherl EJ, Araz Y, Bitar PP, et al. Inflammation-associated adherent-invasive Escherichia coli are enriched in pathways for use of propanediol and iron and M-cell translocation. Inflamm Bowel Dis. 2014;20:1919–1932. doi:10.1097/MIB.0000000000000183.
  • Zhang S, Morgan X, Dogan B, Martin F-P, Strickler S, Oka A, Herzog J, Liu B, Dowd SE, Huttenhower C, et al. Mucosal metabolites fuel the growth and virulence of E. JCI Insight. 2022;7:e157013.
  • Fornelos N, Franzosa EA, Bishai J, Annand JW, Oka A, Lloyd-Price J, Arthur TD, Garner A, Avila-Pacheco J, Haiser HJ, et al. Growth effects of N-acylethanolamines on gut bacteria reflect altered bacterial abundances in inflammatory bowel disease. Nat Microbiol. 2020;5:486–497. doi:10.1038/s41564-019-0655-7.
  • Koh A, De Vadder F, Kovatcheva-Datchary P, From Dietary BF. Fiber to host physiology: short-Chain fatty acids as key bacterial metabolites. Cell. 2016;165:1332–1345. doi:10.1016/j.cell.2016.05.041.
  • Peng K, Xia S, Xiao S, Yu Q. Short-chain fatty acids affect the development of inflammatory bowel disease through intestinal barrier, immunology, and microbiota: a promising therapy? J Gastro Hepatol. 2022;37:1710–1718. doi:10.1111/jgh.15970.
  • Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. the Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell. 2012;48:612–626. doi:10.1016/j.molcel.2012.08.033.
  • Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–455. doi:10.1038/nature12726.
  • Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–450. doi:10.1038/nature12721.
  • Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–139. doi:10.1016/j.immuni.2013.12.007.
  • Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS, Metabolites TM. Short-Chain fatty acids, regulate colonic treg cell homeostasis. Science. 2013;341:569–573. doi:10.1126/science.1241165.
  • Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569:655–662. doi:10.1038/s41586-019-1237-9.
  • Marchesi JR, Holmes E, Khan F, Kochhar S, Scanlan P, Shanahan F, Wilson ID, Rapid WY. Noninvasive metabonomic characterization of inflammatory bowel disease. J Proteome Res. 2007;6:546–551. doi:10.1021/pr060470d.
  • Takaishi H, Matsuki T, Nakazawa A, Takada T, Kado S, Asahara T, Kamada N, Sakuraba A, Yajima T, Higuchi H, et al. Imbalance in intestinal microflora constitution could be involved in the pathogenesis of inflammatory bowel disease. Int J Med Microbio. 2008;298:463–472. doi:10.1016/j.ijmm.2007.07.016.
  • Pace F, Rudolph SE, Chen Y, Bao B, Kaplan DL, Watnick PI, Theis KR. The Short-Chain fatty acids propionate and butyrate augment adherent-Invasive Escherichia coli virulence but repress inflammation in a human intestinal enteroid model of infection. Microbiol Spectr. 2021;9:e01369–21. doi:10.1128/Spectrum.01369-21.
  • Ormsby MJ, Johnson SA, Carpena N, Meikle LM, Goldstone RJ, McIntosh A, Wessel HM, Hulme HE, McConnachie CC, Connolly JPR, et al. Propionic acid promotes the virulent phenotype of Crohn’s disease-Associated adherent-Invasive Escherichia coli. Cell Rep. 2020;30(2297–2305.e5). doi:10.1016/j.celrep.2020.01.078.
  • Pobeguts OV, Ladygina VG, Evsyutina DV, Eremeev AV, Zubov AI, Matyushkina DS, Scherbakov PL, Rakitina DV, Fisunov GY. Propionate induces virulent properties of Crohn’s Disease-Associated Escherichia coli. Front Microbiol. 2020;11:1460. doi:10.3389/fmicb.2020.01460.
  • Zhang S, Dogan B, Guo C, Herlekar D, Stewart K, Scherl EJ, Simpson KW. Short chain fatty acids modulate the growth and virulence of Pathosymbiont Escherichia coli and host response. Antibiotics. 2020;9:462. doi:10.3390/antibiotics9080462.
  • Palmer LD, Skaar EP, Palmer LD, Skaar EP. Transition Metals and. Transition Metals and Virulence in Bacteria. Annu Rev Genet. 2016;50(1):67–91. doi:10.1146/annurev-genet-120215-035146.
  • Golonka R, Yeoh BS, The Iron V-KM. Tug-of-War between Bacterial Siderophores and Innate Immunity. J Innate Immun. 2019;11:249–262. doi:10.1159/000494627.
  • Ellermann M, Gharaibeh RZ, Fulbright L, Dogan B, Moore LN, Broberg CA, Lopez LR, Rothemich AM, Herzog JW, Rogala A, et al. Yersiniabactin-Producing adherent/invasive Escherichia coli promotes inflammation-Associated Fibrosis in Gnotobiotic Il10 −/− Mice. Infect Immun. 2019;87:e00587–19. /iai/87/11/IAI.00587-19.atom
  • Singh V, Yeoh BS, Xiao X, Kumar M, Bachman M, Borregaard N, Joe B, Vijay-Kumar M. Interplay between enterobactin, myeloperoxidase and lipocalin 2 regulates E. Nat Commun. 2015;6:7113.
  • Shaler CR, Parco AA, Elhenawy W, Dourka J, Jury J, Verdu EF, Coombes BK. Psychological stress impairs IL22-driven protective gut mucosal immunity against colonising pathobionts. Nat Commun. 2021;12:6664. doi:10.1038/s41467-021-26992-4.
  • Gracie DJ, Guthrie EA, Hamlin PJ, Ford AC. Bi-directionality of brain–Gut interactions in patients with inflammatory Bowel Disease. Gastroenterology. 2018;154(1635–1646.e3):1635–1646.e3. doi:10.1053/j.gastro.2018.01.027.
  • Gerner RR, Hossain S, Sargun A, Siada K, Norton GJ, Zheng T, Neumann W, Nuccio S-P, Nolan EM, Raffatellu M. Siderophore Immunization Restricted Colonization of Adherent-Invasive Escherichia coli and Ameliorated Experimental Colitis. mBio. 2022;13:e02184–22. doi:10.1128/mbio.02184-22.
  • Flemming HC, Wingender J. The biofilm matrix. Nat Rev Microbiol. 2010;8:623–633. doi:10.1038/nrmicro2415.
  • Flemming H-C, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S. Biofilms: an emergent form of bacterial life. Nat Rev Microbiol. 2016;14:563–575. doi:10.1038/nrmicro.2016.94.
  • Flemming H-C, van Hullebusch ED, Neu TR, Nielsen PH, Seviour T, Stoodley P, Wingender J, Wuertz S. The biofilm matrix: multitasking in a shared space. Nat Rev Microbiol [Internet] 2022 [cited 2022 Sep 21]; Available from]. https://www.nature.com/articles/s41579-022-00791-0
  • Sauer K, Stoodley P, Goeres DM, Hall-Stoodley L, Burmølle M, Stewart PS, Bjarnsholt T. The biofilm life cycle: expanding the conceptual model of biofilm formation. Nat Rev Microbiol. 2022;20:608–620. doi:10.1038/s41579-022-00767-0.
  • Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Spatial Organization LH. Composition of the Mucosal Flora in patients with inflammatory bowel disease. J Clin Microbiol. 2005;43:3380–3389. doi:10.1128/JCM.43.7.3380-3389.2005.
  • Chassaing B, Darfeuille-Michaud A. The σ E pathway is involved in biofilm formation by Crohn’s Disease-Associated Adherent-Invasive Escherichia coli. J Bacteriol. 2013;195:76–84. doi:10.1128/JB.01079-12.
  • Macé K, Vadakkepat AK, Redzej A, Lukoyanova N, Oomen C, Braun N, Ukleja M, Lu F, Costa TRD, Orlova EV, et al. Cryo-EM structure of a type IV secretion system. Nature. 2022;607:191–196. doi:10.1038/s41586-022-04859-y.
  • Costa TRD, Harb L, Khara P, Zeng L, Hu B, Christie PJ. Type IV secretion systems: advances in structure, function, and activation. Mol Microbiol. 2021;115:436–452. doi:10.1111/mmi.14670.
  • Christie PJ, Gomez Valero L, Buchrieser C. Biological diversity and evolution of type IV secretion systems. In: Backert S, Grohmann E, editors. Type IV secretion in Gram-Negative and Gram-positive bacteria. Switzerland: Springer International Publishing; 2017. p. 1–30. cited 2022 Mar 12. Available from: http://link.springer.com/10.1007/978-3-319-75241-9_1
  • Ghigo JM. Natural conjugative plasmids induce bacterial biofilm development. Nature. 2001;412:442–445. doi:10.1038/35086581.
  • Ellermann M, Huh EY, Liu B, Carroll IM, Tamayo R, Sartor RB, Bäumler AJ. Adherent-Invasive Escherichia coli production of cellulose influences iron-induced bacterial aggregation, Phagocytosis, and induction of colitis. Infect Immun. 2015;83:4068–4080. doi:10.1128/IAI.00904-15.
  • Renouf MJ, Cho YH, McPhee JB. Emergent behavior of IBD-associated Escherichia coli during disease. Inflamm Bowel Dis. 2019;25:33–44. doi:10.1093/ibd/izy312.
  • Yang Y, Nguyen M, Khetrapal V, Sonnert ND, Martin AL, Chen H, Kriegel MA, Palm NW. Within-host evolution of a gut pathobiont facilitates liver translocation. Nature. 2022;607:563–570. doi:10.1038/s41586-022-04949-x.