Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Jun 20, 2025; 15(2): 92592
Published online Jun 20, 2025. doi: 10.5662/wjm.v15.i2.92592
Gut virome: New key players in the pathogenesis of inflammatory bowel disease
Helal F Hetta, Yasmin N Ramadan, Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
Helal F Hetta, Division of Microbiology, Immunology and Biotechnology, Faculty of pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
Rehab Ahmed, Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
Hayam Fathy, Department of Internal Medicine, Division Hepatogastroenterology, Assiut University, Assiut 71515, Egypt
Mohammed Khorshid, Department of Clinical Research, Egyptian Developers of Gastroenterology and Endoscopy Foundation, Cairo 11936, Egypt
Mohamed M Mabrouk, Department of Internal Medicine, Faculty of Medicine. Tanta University, Tanta 31527, Egypt
Mai Hashem, Department of Tropical Medicine, Gastroenterology and Hepatology, Assiut University Hospital, Assiut 71515, Egypt
ORCID number: Helal F Hetta (0000-0001-8541-7304); Rehab Ahmed (0000-0003-2476-469X); Yasmin N Ramadan (0009-0008-7374-9334); Hayam Fathy (0000-0001-5289-303X); Mohammed Khorshid (0000-0002-8466-0940); Mohamed M Mabrouk (0000-0002-2463-1347); Mai Hashem (0000-0002-7877-0094).
Author contributions: Hetta HF, Ahmed R, Ramadan YN, Fathy H, Khorshid M, Mabrouk MM, and Hashem M participated in the study design, prepared the tables and figures, wrote the manuscript, performed some of the analyses, and revised the manuscript; All authors read and approved the final manuscript.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Mai Hashem, MBChB, MD, MHSc, Lecturer, Department of Tropical Medicine, Gastroenterology and Hepatology, Assiut University Hospital, Assiut University Campus, Assiut 71515, Egypt. mayahashem@yahoo.com
Received: January 30, 2024
Revised: May 28, 2024
Accepted: July 23, 2024
Published online: June 20, 2025
Processing time: 301 Days and 22 Hours

Abstract

Inflammatory bowel disease (IBD) is a chronic inflammatory illness of the intestine. While the mechanism underlying the pathogenesis of IBD is not fully understood, it is believed that a complex combination of host immunological response, environmental exposure, particularly the gut microbiota, and genetic susceptibility represents the major determinants. The gut virome is a group of viruses found in great frequency in the gastrointestinal tract of humans. The gut virome varies greatly among individuals and is influenced by factors including lifestyle, diet, health and disease conditions, geography, and urbanization. The majority of research has focused on the significance of gut bacteria in the progression of IBD, although viral populations represent an important component of the microbiome. We conducted this review to highlight the viral communities in the gut and their expected roles in the etiopathogenesis of IBD regarding published research to date.

Key Words: Inflammatory bowel disease; Pathogenesis; Gut virome; Bacteriophage; Eukaryotic viruses

Core Tip: Inflammatory bowel disease (IBD) is a chronic multifactorial inflammatory disease involving the gastrointestinal tract. The exact etiopathogenesis is unknown, but it’s believed that gut microbiome dysbiosis is a cornerstone in triggering disease progression. The gut virome forms a significant part of the gut microbiome and participate in health and disease conditions. Until 2015, researchers paid little attention to their role in IBD. Subsequently, numerous studies have followed this line of inquiry, using advanced techniques to clarify this role. Herein, we emphasize the viral populations in the gut and their predicted roles in the etiopathogenesis of IBD based on current studies.



INTRODUCTION

Inflammatory bowel disease (IBD), including Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic condition characterized by chronic inflammation of the gastrointestinal tract (GIT)[1]. The precise etiology of IBD is complex and still not fully understood. However, studies have revealed that the onset and course of IBD are controlled by a variety of factors, including the interaction between environmental factors (e.g., intestinal microbiota) and the host immune response in genetically susceptible people[2-6]. After birth and in the early days of life, the gut microbiota begins to colonize the GIT, where they coexist in an equilibrium process and actively interact with the host[7]. In healthy settings, the composition of microbiota changes until adulthood, when it becomes more stable[8]. In particular, the gut microbiota maintains the integrity of the gut barrier, promotes the generation of nutrients (e.g., short-chain fatty acids [SCFAs] and vitamins), regulates the immunological response, and participates in the metabolism of drugs and nondigestible food, and defense against pathogenic organisms[9,10]. Typically, gut microbiota consists of bacteria, viruses, fungi, and archaea. Bacteria have received the most attention from these microbes and have been associated with developing mucosal immunity and reducing mucosal inflammation[11-14]. An abnormality in one of these immunological pathways can have a negative impact on IBD development. For instance, alterations in the function of the bacterial microbiome or a decrease in Bacteroidetes and Firmicutes levels and an increase in less prevalent bacterial species (spp.) have all been linked to IBD[15]. Nonbacterial elements of the gut microbiota have been neglected in previous studies for a variety of reasons, including their low absolute prevalence in the intestinal microbiota of humans and a scarcity of competent and specific diagnostic methods for nonbacterial genome analysis[16].

Early studies defining the gut microbiota focused on culturing bacteria, which had little success since only a tiny fraction of gut microbes can be cultivated[17]. Following that, in the early 2000s, next-generation sequencing technology emerged and allowed scientists to investigate the diversity of gut microbiota. This scientific advancement led to the emergence of the “microbiome” era, which aims to study the whole microbial genomes, paving the way for the development of the subfield of “virome research”[18]. The gut virome is still a little-studied subsection of the whole microbiome despite this significant development[19]. Regardless of the lack of representation, several publications have demonstrated that a disturbed gut virome is linked to several illnesses including type 1 and type 2 diabetes[20,21], cystic fibrosis[22], obesity[23,24], graft vs host disease[25], acquired immunodeficiency syndrome[26], colorectal cancer[27], malnutrition[28], liver diseases[29], severe acute respiratory syndrome coronavirus 2[30], as well as IBD[31].

This review provides deep insights into gut virome dysbiosis and its role in the etiopathogenesis of IBD.

AN OVERVIEW OF THE GUT VIROME

The GI system has a complex ecosystem, including bacteria, viruses, fungi, and protozoans. The overall GI microorganism communities and their constituent genes are known as the gut microbiome[32]. The gut microbiome plays a key role in developing and maintaining homeostasis and a balanced immune system through interactions with epithelial and immune cells and regulating metabolic processes (such as SCFAs and bile acids)[33-35]. Viruses form a significant part of the gut microbiome and participate in maintaining homeostasis[36].

The two main forms of viruses in the gut microbiome are phages, which infect bacteria, and viruses, which infect eukaryotic cells (such as human cells). Although both kinds have been observed in the human GIT, phages account for the vast majority of viral spp.[37] Both forms either contain DNA or RNA (single or double strand) as genetic material[38]. A phage enters its cellular host and uses its machinery to start its own reproduction process. There are two major lifecycles that characterize this process: lytic or lysogenic cycle[39]. The lytic cycle comprises attachment, entry, replication, and creation of virions, which are mainly completed through lysis of the host cell (Figure 1)[40]. The lysogenic cycle involves the formation of extrachromosomal plasmids in the cytoplasm or the integration of phage genetic material into the host genome[40]. The lysogenic cycle allows viruses to remain latent (as prophages), which ensures that the genetic material will be passed on to cellular progeny during cell division[37,41]. Induction of prophage happens either naturally at a low rate or is activated by outside stresses, initiating the DNA damage response or SOS response (Figure 1)[41,42]. Moreover, Erez et al[40] discovered the “arbitrium system,” a phage-specific communication mechanism that enables the phages to detect their levels in the surroundings and choose whether to start the lytic or lysogenic cycle. Using this approach, phages lysogenize the host at high arbitrium concentrations and lyse the host at low arbitrium concentrations[40]. Prophage serves as a reservoir for phage-encoded genes that the bacterial host may acquire[43]. This genetic reservoir may have genes that increase stress tolerance and immunity, promote virulence and biofilm production, and provide metabolic and antibiotic resistance[44-48]. As a result, these acquired bacterial activities might be advantageous (as boosting immunity) or damaging (as virulence factors) to the human host[49]. For instance, prophages can protect bacterial host cells from subsequent infection from closely related phages[49]. Moreover, prophages have the ability to disseminate virulence components that turn some commensal gut microbiota into pathogenic ones[46,50]. For instance, phages from the Inoviridae family that encode cholera toxin can incorporate with the bacterial host, transport toxin genes, and produce dangerous organisms[50,51].

Figure 1
Figure 1 Two major lifecycles of bacteriophage (lytic and lysogenic).
GUT VIROME IN NORMAL HEALTHY CONDITIONS

Each human has a large number of viruses, approximately 1013 particles per person, the majority of which are located in the gut[52-54]. According to growing data, the gut microbiome is initially quite basic, changes quickly during the first days after childbirth, and eventually becomes more diversified and stable over time[55-58]. Breitbart et al[59] conducted the first investigation documenting the gut phage population in fresh fecal samples in newborns. According to their investigation, the meconium, a newborn’s initial fecal excretion, failed to include any virus-like particles (VLPs) when examined by a direct epifluorescence microscope. On the other hand, towards the end of the first week, 108 VLPs per gram of moist feces were found[59].

Furthermore, in 2015, two additional studies revealed that the gut phage population exhibited considerable changes throughout the first 2 and 2.5 years of life, respectively[60,61]. Lim et al[60] documented the existence of the Microviridae family in the dominant phages in addition to the Caudovirales class, as well as a shift from Caudovirales to Microviridae at the first 24 mo of age. Additionally, they discovered that the abundance as well as diversity of intestinal phages were maximized in the first 4 d of life and subsequently declined as individuals aged[60].

Members of the phage classes Malgrandaviricetes (spherical single-strand DNA [ssDNA]) and Caudovirales (tailed double-strand DNA) make up the biggest known populations of viruses living in a healthy human GIT[57,62]. Caudovirales are believed to infect Bacteroidetes, Firmicutes, Verrucomicrobia, Actinobacteria, and Proteobacteria, while Malgrandaviricetes are believed to infect Enterobacteria or intracellular microorganisms (such as Spiroplasma, Chlamydia, and bdellovibrio)[63,64]. The crAss-like phage, a newly identified monophyletic clade within the Caudovirales class, is thought to be the most abundant phage in the healthy human gut[62,65,66]. After the identification of crAss-like phages, several other prevalent and common viral clades, including Flandersviridae, Gubaphage, giant Lak phages, and LoVEphage, were discovered[67-69]. Small circular ssDNA viruses, such as Anelloviridae and Caudovirales, are among the most common eukaryotic viruses[62,70]. It was reported that Anelloviruses are not particularly common, but they form a very diverse and common eukaryotic viral class at the beginning of life and are reduced gradually as the microbiome matures[63]. Plant viruses are another type of eukaryotic virus that is typically observed in elevated concentrations in the healthy human GIT[71]. They are often gained by food and passed to the GIT[62,72].

Viral complexity is defined by substantial interindividual variations or notable uniqueness of viral contigs[62,73]. While people significantly differ from one another, a person’s virome can remain quite constant over time, as demonstrated by low intraindividual variance[74,75]. Generally, it is believed that the healthy gut microbiota is a diverse ecosystem, and any dysbiosis or imbalance in this ecosystem is frequently linked to the progression of diseases[76] such as IBD[6,77,78], irritable bowel syndrome[79-81], and colorectal cancer[82-84]. Although the patterns of diversity in the healthy gut virome are still out of reach, it is believed that they have a positive impact on the diversity of the microbiome[85].

ENVIRONMENTAL AND HOST FACTORS THAT AFFECT THE GUT VIROME

Many factors can affect and shape the gut virome. One of these is anthropometric factors that measure the physical properties of the host such as height, weight, age, and body mass index, among others[62,86,87]. Other factors can be divided into a number of main groups, including nutrition and its relationship to stool uniformity, lifestyle, and physical activity, diseases and medications, as well as geographical location[24,74,86,88-90]. As mentioned above, the diversity of the gut virome reaches its maximum level in the first days after birth and decreases with age[60]. Also, it is thought that people’s dietary habits impact the type of viruses in their GIT[74,91,92]. For example, consuming coffee and dairy products and consuming fruit have a positive association with the diversity of the gut virome and affect the Shannon diversity index and Bristol Stool Score[74,86,93,94]. In addition, consumption of high quantities of fats is linked to a low proportion of Caudovirales phages and a large proportion of Malgrandaviricetes phages, as well as reduced lysogenic capacity[92]. Several investigations have shown that the medications might activate prophages inside their hosts, modifying the virome of the gut, and subsequently, the life cycle[95,96]. A complex equilibrium between lytic and lysogenic phages is therefore believed to exist in a healthy condition[57]. For instance, a lysogenic lifestyle results in a higher bacterial cell number, but activation of lytic activity results in a lower bacterial cell number[57,97]. Higher microbial cell numbers probably give phages an easier means to replicate their incorporated genomes via bacterial replication instead of lysing the bacterial host[97]. Caudovirales phages, which are mainly lysogenic, are abundant in the early stages of the development of the neonatal gut virome, but as time goes on, Malgrandaviricetes phages, which are obligatorily lytic, become more prevalent[60,62,98]. This means that a reduced lysogenic and greater lytic capacity of the intestinal virome occur toward adulthood[63].

In conclusion, lysogenic and lytic phages are in a dynamic equilibrium in a healthy adult gut, and some factors can disrupt this equilibrium and encourage the induction of prophages.

PATHOPHYSIOLOGY OF IBD WITH THE ROLE OF THE GUT VIROME IN IBD

Research conducted over the past 30 years on human intestinal tissue and in vivo mouse models has revealed that intestinal homeostasis, which governs the host-microbiome interaction, is largely dependent on the integrity of the epithelial barrier, host defense mechanisms, immunological modulation, and tissue repair. Any disruption of these pathways or the cytokine networks that regulate them can result in IBD[5,99,100]. IBD is a multifactorial disease. The pathophysiology may be initiated by dysbiosis in the gut ecosystem as a result of some environmental or genetic factors[101]. Subsequently, dysbiosis triggers several inflammatory pathways[33,102,103] (Figure 2)[104]. However, the exact association between dysbiosis and inflammation in IBD is yet to be understood. Whether dysbiosis is the cause of inflammation or the inflammation leads to dysbiosis, the final result is the coexistence of dysbiosis and inflammation and the progression of IBD[104].

Figure 2
Figure 2 Role of gut microbiome in health condition and inflammatory bowel disease progression. In a healthy state, a balanced microbiome helps maintain homeostasis and the production of beneficial metabolites that build tight junctions and keep gut epithelium barrier integrity. On the other hand, gut microbiome dysbiosis participates in damaging tight junctions as well as dysregulating the gut barrier. This enhances the interaction of pathogens with gut epithelium and increases their penetration to the gut lumen with subsequent stimulation to immunological response and immune cells and overproduction of proinflammatory cytokines and progression of inflammatory bowel disease. PBAs: Primary bile acids; SBAs: Secondary bile acid; SCFAs: Short-chain fatty acids; IBD: Inflammatory bowel disease.

The onset and progression of IBD are influenced by several critical risk factors including genetics[105,106], diet[107,108], smoking[109], medicines, stress, mental health, and others[110]. Up to 12% of cases indicate a family history of illness, making genetics the most significant known risk factor. Additionally, diet and dietary habits have significant effects on the initiation of IBD[107]. More precisely, a low-fiber diet is thought to switch the gut microbiome from digesting fiber-derived glycans to digesting mucus-derived glycans, destroying the mucous protective layer of the gut and enhancing pathogen penetration with subsequent activation of inflammatory cascades[111]. These inflammatory responses are characterized by gut microbiome dysbiosis, including virome dysbiosis. Gut virome dysbiosis includes the reduction of Microviridae and crAss-like phages as well as the propagation of Caudovirales and perhaps other phages with lysogenic potential[31,112,113]. Regarding eukaryotic viruses, it has been revealed that patients with IBD have higher prevalence rates of particular viral families (e.g., Herpesviridae and Anelloviridae) than normal controls[31,112,114,115].

IMPLEMENTATION OF PHAGE IN IBD

After reviewing the literature, we found that gut phages may influence IBD pathogenesis by three mechanisms: (1) Change of gut phage community; (2) Modulation of gut microbial population; and (3) Modification of the local immune response.

Change of gut phage community

Regarding the alteration of the gut phage community, the majority of investigations depend on metagenomic sequencing of stool samples and intestinal biopsies. Variations among normal people and patients with IBD or experimental models have been discovered (Table 1).

Table 1 Overview of the alteration in the gut phage ecosystem in patients with inflammatory bowel disease and animal models.
Disease
No. of patients included in the study
Sample type
Interpretation of result
Ref.
CD19BiopsiesCD patients had considerably more VLPs than normal controls[116]
CD6Ileal biopsies, colonic biopsies, gut wash samplesA significant excess of phages in biopsies and gut washes, Bacteroides phages (B10-8 and B124-14) were most predominant, and the composition of Mycobacterium phage differed between CD patients and controls in ileum tissue samples[117]
CD20Stool samples, biopsiesPhage counts in stools were three times greater than in biopsies, CD patients had higher levels of Alteromonadales and Clostridiales phages[114]
IBD10Colonic biopsiesPhages make up the bulk of the DNA viruses within the virome, about 50% of the phages were connected to the bacterial strains found in the colon specimens[118]
UC and CD(42 for UC) and (18 for CD)Stool samplesPatients with IBD had a considerable increase in Caudovirales phages, and virome community in UC and CD patients were disease and cohort-specific[31]
UC and CD(5 pt. for UC) and (7 pt. for CD)Stool samplesCaudovirales phage proportions in patients with IBD and normal controls were greater than Microviridae phage proportions. However, the Caudovirales phages were more prominent in CD than UC but not in controls. On the other hand, control persons had a larger diversity of Microviridae phages than CD patients, but not UC patients[119]
UC97Rectal mucosaCaudovirales phages were more abundant in UC cases compared to normal controls, but with lower richness, diversity, and balance, and UC patients’ mucosa had much more Enterobacteria and Escherichia phages than healthy controls[113]
UC and CD(42 pt. for UC) and (27 pt. for CD)Stool samplesA stable virulent core virome is associated with a healthy gut and switched from a lysogenic to lytic cycle in temperate phages may be related to CD[112]
CD5proximal and distal colonic wash samplesConsiderable interpatient diversity and little, but significant, intrapatient variations between various regions[120]
(VEO) IB45Stool samplesNo detectable difference in the overall number of VLPs among VEO-IBD patients and normal controls, but the Caudovirales vs Microviridae ratio is larger in the VEO-IBD patients than in the controls[121]
UC and CD(38 pt. for UC) and (65 pt. for CD)Stool samplesThe prevalence of phages varied among patients with IBD and normal controls as well as the components of the temperate phage population were extremely distinctive to each individual. Moreover, compared to normal controls, active UC patients had a higher prevalence of temperate phages infecting Bacteroides thetaiotaomicron and Bacteroides uniformis[122]
IBD455Stool samplescrAss-like phageome of the human gut has remained largely stable for 4 yr and individuals with IBD had lower levels of gut crAss-like phages[65]
CD19Stool samplesCD patients had a considerably higher prevalence of crAss-like phages as well as no difference in the richness and evenness of the gut virome among CD patients and controls, but there was a substantial difference in the virome’s overall structure[123]
Colitis3 from C57BL/6 miceStool samplesThe intestinal phage populations were altered and shifted to dysbiosis in the mice model, and a decrease in the variety of the phage community, such as Clostridiales phages during colitis[124]

In 2008, Lepage et al[116] published the first study connecting phages to IBD. They used epifluorescence and electron microscopy to compare populations of VLP in biopsies from patients with CD and healthy controls. They discovered that patients with CD had considerably more VLPs compared to healthy people[116]. This opened the door for further research to demonstrate the gut virome in different IBD subtypes and shed light on the role of the virome in the progression of IBD. Wagner et al[117] conducted a study on pediatric patients with CD to compare the alteration in phage population in GI biopsies from different sites and gut wash between patients and control individuals. They collected tissue biopsies from the ileum and colon as well as gut wash and analyzed them through metagenomic analysis. They found a significant excess of phages in biopsies and gut washes of pediatric CD patients compared to healthy controls. Furthermore, they discovered that the Bacteroides phages (B10-8 and B124-14) were the most predominant, and the composition of Mycobacterium phage differed between CD patients and controls in ileum tissue samples[117]. Further metagenomics examination of colonic specimens revealed that about 50% of phages were connected to the bacterial strains found in the colon specimens[118]. Subsequently in 2015, Pérez-Brocal et al[114] demonstrated variations in the gut bacteriome and virome communities in various types of specimens from adult patients with CD at various stages. They discovered that the phage counts in stools were three times greater than in biopsies and that the bacterial community rather than the viral populations are a better predictor of an individual’s illness status. Also, they discovered that individuals with CD had higher levels of phages infecting the bacterial orders Alteromonadales and Clostridiales, including Clostridium acetobutylicum spp. as well as Retroviridae family[114]. In the same year, Norman et al[31] established a metagenomic analysis to demonstrate the differences in gut phage populations in stool samples among UC and CD patients vs healthy controls. They showed that, compared to healthy groups, patients with IBD had a considerable increase in Caudovirales phages. Additionally, the CD and UC patients’ gut phage community was disease and cohort-specific[31]. Later, in 2019, Fernandes et al[119] examined the virome of fecal samples in children with CD, UC, and healthy controls of the same age. The result showed that Caudovirales phage proportions in both patients with IBD and normal controls were greater than Microviridae phage proportions. However, the Caudovirales phages were more prominent in CD than UC but not in controls. On the other hand, the control group showed a larger diversity of Microviridae phages than patients with CD, but not those with UC[119]. Moreover, another study by Zuo et al[113] identified the virome communities of the mucosa of patients with UC. According to their investigation, Caudovirales phages were more abundant in UC cases compared to normal controls but had lower richness, diversity, and balance. They also discovered that the mucosa of patients with UC had much more Enterobacteria and Escherichia phages than healthy controls[113]. Interestingly, Clooney et al[112] employed a whole-virome sequencing technique to re-analyze previously published data and provide extensive insights into the activity of the gut virome and its possible involvement in IBD[112]. They found that a stable virulent core virome is associated with a healthy gut, and switching from a lysogenic to lytic cycle in temperate phages may be related to CD[112]. A new virome sequencing analysis using pediatric CD patients’ proximal and distal colonic wash samples revealed considerable inter-patient diversity and little but significant intra-patient variations among different regions[120]. In another study, Liang et al[121] evaluated the dynamics of the virome in stool samples collected from children classified with very early onset (VEO) IBD, defined as IBD with onset prior to the child’s sixth birthday. They found that there is no detectable difference in the overall number of VLPs among VEO-IBD patients and normal controls but that the Caudovirales vs Microviridae ratio is larger in VEO-IBD patients than in the controls[121]. By utilizing whole-metagenome shotgun sequencing data, Nishiyama et al[122] showed the ecological composition of the temperate phage population in the human gut. They discovered that the prevalence of phages varied among patients with IBD and normal controls, and the components of the temperate phage population were extremely distinctive to each individual. Moreover, compared to normal controls, patients with active UC had a higher prevalence of temperate phages infecting B. thetaiotaomicron and B. uniformis[122]. In the most recent study, Gulyaeva et al[65] performed metagenomic sequencing on feces specimens collected from 1950 individuals, to investigate the vital function and diversity of crAss-like phages in clinical cohorts and human populations. They found that the crAss-like phageome of the human gut remained stable for 4 years and that individuals with IBD had lower levels of gut crAss-like phages[65]. Furthermore, Imai et al[123] analyzed stool samples collected from Japanese patients with CD using shotgun metagenomic sequencing. In contrast, they found that patients with CD had a considerably higher prevalence of crAss-like phages as well as no difference in the richness and evenness of the gut virome among CD patients and controls, but there was a substantial difference in the overall structure of the virome[123].

Animal studies additionally represent an essential tool for investigating the functions of intestinal phages in the pathophysiology of IBD. Duerkop et al[124] reported that in an animal model of colitis, the intestinal phage populations were altered and shifted to dysbiosis[124]. Also, they noticed a decrease in the variety of the phage community, such as Clostridiales phages, during colitis.

In summary, recent studies employed metagenomic sequencing and bioinformatic analysis to describe fecal and mucosal phage ecosystems. Most studies found that Caudovirales phages were more frequent and less diverse in patients with CD and UC than normal controls. However, different results were found in a recent study that showed no substantial variation in the number of intestinal phages between patients with IBD and normal controls[121]. Additionally, the abundance of crAss-like phage and Microviridae was decreased. Some investigations also revealed changes in specific phages, such as elevated levels of Alteromonadales and Clostridial phages in CD patients, elevated levels of Escherichia and Enterobacteria phages in UC mucosa, and decreased levels of Clostridial phages during colitis.

Modulation of the gut microbial population through bacteriophages

Virulent phages, which have the ability to lyse the bacterial host cell, are frequently identified in the GIT of patients with IBD[112]. It has been demonstrated that the invasion of phage to its bacterial host leads to modification and alteration in bacterial community with subsequent change in the abundance of certain spp.[125]. Researchers have shown that individuals with CD and UC exhibit dysbiosis in their gut microbiome, which is characterized by reduced diversity, increased hazardous proteobacteria (e.g., E. coli and Fusobacteria), and decreased beneficial Firmicutes (e.g., Clostridium clusters IV and XIVa, Faecalibacterium prausnitzii, and Rumininococci)[126-128]. Additionally, Nishiyama et al[122] found a significant increase in the prevalence of phages infecting B. thetaiotaomicron and B. uniformis, as well as a reduction in the population of their bacterial host. Considering the aforementioned studies, we conclude that there is a link between the abundance of phage and its bacterial host population. Germ-free (GF) animals are the ideal model for investigations related to the gut microbiome since they don’t have any microbial colonization in their guts[129,130]. In GF murine models, a recent investigation revealed that phage invasion directly affects vulnerable bacteria, with subsequent cascade affecting other bacterial spp. and gut metabolome[131,132].

Other than virulent phages, temperate phages can affect the viability and diversity of gut bacteriome[133]. For instance, temperate phages significantly increase the genetic variation of bacteria via horizontal gene transfer and increase the mutation rates[46,133-135]. Moreover, the induction of latent prophage through environmental stressors may activate its lytic cycle and decrease the number of bacterial hosts. In a metagenomic study conducted by Cornuault et al[136], they found a greater abundance or quantity of phages infecting F. prausnitzii in feces samples from patients with IBD in comparison to normal controls[136]. While less F. prausnitzii abundance has been demonstrated in patients with IBD, they concluded that phages might exacerbate this reduction of F. prausnitzii[136].

In summary, the relevant information is still inadequate, and theories about how phages directly or indirectly affect bacterial populations are still out of reach. So further investigations are required to fully understand the complex phage-bacteria interactions in IBD.

Modification of the local immune response through bacteriophages

After prophage induction, a process known as phage-mediated lysis describes the positive feedback inflammatory response between phage induction and gut inflammation-begins[133,137]. In this situation, intestinal inflammation induces the production of stressors by enterocytes, such as reactive oxygen species and reactive nitrogen species, which cause the host bacteria to respond to stress (SOS response)[138]. Increased bacterial host cell lysis is followed by a rise in pathogen-associated molecular patterns (PAMPs) (such as lipopolysaccharides and bacterial DNA) that activate more enterocyte receptors[133,137]. This leads to activation of a positive feedback inflammatory response and dysregulation of the immune system (Figure 3). Additionally, in the presence of a thin lining mucous layer and disrupted tight junction, large amounts of PAMPs can penetrate gut epithelium and activate Toll-like receptors (TLRs) and other immune cells located on gut epithelium[31,139-141]. As a result, inflammatory pathways are activated, resulting in increased generation of pro-inflammatory cytokines and decreased generation of anti-inflammatory cytokines[142-145] (Figure 4). A recent in vivo investigation conducted in GF mice showed upregulation in both innate and acquired immunity after administration of a phage cocktail. The results demonstrated a considerable increase in overall CD8+ and CD4+ T cells, in addition to interferon gamma (IFN-γ)-producing T helper 1 cells[146]. Furthermore, an in vitro study indicated that the detection of phage DNA by dendritic cells triggers the generation of IFN-γ through a TLR9-dependent mechanism[147].

Figure 3
Figure 3 Activation of positive feedback inflammatory response through induction of latent prophage and lysis of bacterial host cell. (1) Intestinal inflammation induces gut mucosa to generate stressors (like reactive oxygen species [ROS] and reactive nitrogen species [RNS]); (2) Production of stressors aggregates the stressor response (SOS) in the bacterial host cell (SOS reaction) and redox imbalance; (3) This imbalance leads to damage of bacterial DNA and induction of latent phage; (4) Switch to lytic cycle with subsequent bacterial cell rupture; (5) Accumulation of pathogen-associated molecular patterns (PAMPs) as lipopolysaccharide (LPS) and DNA, that results from bacterial lysis; (6) PAMPs activate receptors in the gut mucosa and stimulate the production of more stressors.
Figure 4
Figure 4 Modification of local immune response through bacteriophage. An inflamed gut is characterized by microbiome imbalance including virome. An imbalanced virome is distinguished by an expansion of Caudovirales phages and lysogenic lifecycle as well as a decline in the relative abundance of the Microviridae family and crAss-like phage. Additionally, in the presence of a thin mucus lining or broken tight junction, microbial antigens (like viral antigens) can penetrate the intestinal epithelium, activate Toll-like receptor (TLR), upregulate pro-inflammatory cytokines production, and dysregulate anti-inflammatory cytokines production. IL: Interleukin; RNS: Reactive nitrogen species; ROS: Reactive oxygen species; TGF-β: Transforming growth factor beta; TNF-α: Tumor necrosis factor alpha.

On the other hand, phages may play a significant role in protecting the intestinal barrier against bacteria and provide non-host-derived immunity[148]. Phages can stick to the mucus layer of the gut and reduce the colonization of pathogens. This adhesion was controlled by interactions between immunoglobulin-like domains, which are present on phage capsid proteins, and glycan residues, which are found in mucin glycoprotein[148].

In summary, little information is currently known about gut phages’ impact on IBD through immunological modulation. Therefore, there is an urgent need for more research on how gut phages and the immune system interact in IBD.

IMPLEMENTATION OF EUKARYOTIC VIRUSES

Early in life, eukaryotic viruses begin to colonize the gut mucosa. These viruses are members of the Anelloviridae, Adenoviridae, Picornaviridae, Picobirnaviridae, Astroviridae, and Parvoviridae families, and they become more diverse with age[60]. Such viruses may cause pathological changes or may remain dormant in healthy persons for many years, exerting significant benefits[141,149-151]. Eukaryotic virome dysbiosis, such as phage dysbiosis, has been linked to IBD pathophysiology[152-154] as eukaryotic-targeting viruses incorporate their genetic element into the human genome and can affect the physiological condition of enterocytes[141,151,154]. Patients with UC had greater levels of the eukaryotic Pneumoviridae family than controls, according to a metagenomic study involving a large cohort of UC patients. However, control individuals had higher levels of the eukaryotic Anelloviridae family[113]. On the other hand, another investigation demonstrated that patients with UC and CD had greater levels of the Herpesviridae family than normal controls[118].

Epstein-Barr virus and cytomegalovirus are the most studied eukaryotic viruses that may cause intestinal inflammation[150]. However, their role in the pathophysiology of IBD has yet to be fully understood, as their reactivation may be brought on by immunosuppressive or stressful situations that are prominent in patients with IBD, making them more likely to serve as bystanders than true disease-causing factors.

Norovirus infection was found to be a significant colitogenic factor, significantly dependent on the presence of gut microbiome, in the interleukin 10-deficient mouse model of spontaneous colitis[155]. Likewise, IBD-susceptibility gene Atg16L1HM mouse models have shown that Norovirus infection leads to the progression of intestinal inflammation[156]. Hence, it appears that the development of colitis is accelerated by a synergistic interaction between genetic makeup and Norovirus infection as a trigger of intestinal inflammation.

These aforementioned investigations specifically focused on enterotropic viruses, which are often restricted to the GI system. On the other hand, a recent investigation using metatranscriptomic processes revealed that some eukaryotic RNA viruses with a physiological hepatic tropism were found in the gut mucosa of patients with IBD[151]. In a recent study, Massimino et al[157] discovered how the hepatitis B virus X protein, a virome-associated protein encoded by the Orthohepadnavirus genus, contributes to the pathogenesis of UC.

In summary, previous findings have indicated a link between these eukaryotic virus families and IBD pathogenesis, and more research is urgently required to demonstrate their roles in producing chronic intestinal inflammation.

CHALLENGES AND LIMITATIONS AND FUTURE TRENDS IN VIROME DESCRIPTION

Analysis of the gut virome has been neglected due to the difficulty in producing an in vitro composite culture environment that would support the simultaneous development of different microorganisms[158]. It is still challenging to recognize and categorize viral DNA in microbiological samples. Viral spp. are difficult to classify into closely related spp. due to their extraordinarily high diversity, low gene content, and quick acquisition of mutations[159]. Moreover, it is impracticable to sequence viral DNA using a targeted amplicon-like strategy as there are non-common genes that might be utilized as markers for identification[160]. Unfortunately, it can be challenging to cultivate viruses as well. Because viruses are parasitic and depend on host cells for energy and multiplication, these hosts must also be discovered and cultivated. Additionally, many GIT microorganisms cannot be cultivated, making it problematic to culture their related viruses[115].

Metagenomic analysis of the virome may appear to be a difficult process, but various approaches might help with this issue. For instance, before sequencing, viral particles from a microbiome sample can be separated and purified using size selection by centrifugation, filtration (using 0.2-μm to 0.45-μm filters), and particle precipitation using polyethylene glycol[161]. Despite these helping approaches, the metagenomic technique possesses great limitations, such as dependence of the result on the degree of fragmentation of viral genome as well as analysis of DNA sequence only and ignoring RNA[162]. Although revolution in -omics approaches, such as metagenomics, metataxonomics, and metatranscriptomics, share common limitations[152]. (1) They must depend on reliable databases that provide information on the various genomes and their explanation; otherwise, the analysis will be challenging and may miss some crucial information[158]; (2) Studies must be carried out on purified RNA and DNA specimens, and occasionally, the yields are insufficient to cover poorly represented communities. In addition, residual host DNA and RNA molecules may persist in the sample after purification, leading to false outcomes[158]; (3) The sequencing depth must be very high to obtain accurate outcomes, particularly for metatranscriptomics, which may be expensive[163]; and (4) The present statistical method is constrained by the concept that the predictor variables are independent of one another and do not consider the complexity of the biological ecosystem[158].

The use of more modern computational techniques, such as VIP and VirFinder, which offer workflows to map, filter, and detect viruses from metagenomic sequences[164,165], as well as METAVIR, an online library for identifying viral genes from metagenomic data[166], can make the understanding of human virome easier. Future gut virome investigations should include approaches like tracking viral protein exacerbation[167] or host DNA reduction, as well as high-throughput sequencing of the microbiome in patient samples[159]

COMMUNITY TYPING AS A NEW APPROACH TO DESCRIBING GUT VIROME

The idea of community typing, also known as “enterotyping” was first developed in bacterial studies to simplify the complexity and categorize the diversity of the gut microbiome[168]. The Dirichlet Multinomial Mixture approach is used for community typing, which is based on probability-based modeling and takes into account particular microbiome data properties such as relative scarcity[169]. With this technique, samples from the same community (those with comparable bacterial abundance patterns) are classified into microbial configurations without stating any assumption about the underlying separate character of the strata[169]. These techniques reliably divided the gut microbiome into the 4 enterotypes Ruminococcus, Bacteroides 1, Bacteroides 2 (Bact2), and Prevotella and found several connections to the abovementioned risk factors, including diet and illnesses[170-173].

Due to the massive insights and enterotyping helping in the understanding of the microbiome of the human gut[171,174,175], it has been hypothesized that viral community typing might be a valuable method to pursue knowledge of the gut virome as well. Regarding this idea, Song et al[176] analyzed many published data and found that most people could be sorted into two viral community types depending on their gut virome; however, they were unable to identify their taxonomical makeup because of the elevated incidence of viral dark matter. Additionally, it has been demonstrated that the gut virome of patients with IBD existed in two viral community types: community type CrM, which includes either crAss-like phage and Malgrandaviricetes, or community type CA, which includes Caudovirales phages[177]. Moreover, the community type CA was linked to reduced virome diversity, dysbiosis in the Bact2-enterotype, and active illness, demonstrating the clinical potential of these community types[177,178].

In summary, viral community typing has great promise as a future strategy for discovering alterations in viral composition in health and illness.

CONCLUSION

IBD is a multifactorial chronic inflammatory disease involved in GIT. IBD is divided into two subtypes: UC and CD. The exact etiopathogenesis is still unknown, but the researchers are doing their best to remove this ambiguity. Most researchers focus on the role of gut bacteriome in the etiogenesis of IBD and ignore other microorganism communities as viruses. In 2015, Norman and partners conducted the first investigation revealing the role of gut virome dysbiosis in IBD. Since then, many studies have been conducted with evolution in novel approaches to describe virome dysbiosis and its role in disease and health conditions. In this review, we give an overview of gut virome and its role in normal health conditions. Further, we give deep insights into the implementation of gut virome in IBD pathogenesis regarding the role of both bacteriophages and eukaryotic viruses. Finally, we describe the challenges and limitations in describing gut virome and how the appearance of novel approaches as community typing opens the door for further research to understand the role of gut virome in disease states, including IBD.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medical laboratory technology

Country of origin: Egypt

Peer-review report’s classification

Scientific Quality: Grade D, Grade C

Novelty: Grade B, Grade C

Creativity or Innovation: Grade C, Grade C

Scientific Significance: Grade B, Grade B

P-Reviewer: Day AS S-Editor: Liu H L-Editor: Filipodia P-Editor: Yuan YY

References
1.  Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet. 2007;369:1641-1657.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, Vavricka SR, Fiocchi C. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15:39-49.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Turpin W, Goethel A, Bedrani L, Croitoru Mdcm K. Determinants of IBD Heritability: Genes, Bugs, and More. Inflamm Bowel Dis. 2018;24:1133-1148.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev. 2014;13:3-10.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474:298-306.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Sartor RB, Wu GD. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology. 2017;152:327-339.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Drago L, Valentina C, Fabio P. Gut microbiota, dysbiosis and colon lavage. Dig Liver Dis. 2019;51:1209-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Zwittink RD, van Zoeren-Grobben D, Martin R, van Lingen RA, Groot Jebbink LJ, Boeren S, Renes IB, van Elburg RM, Belzer C, Knol J. Metaproteomics reveals functional differences in intestinal microbiota development of preterm infants. Mol Cell Proteomics. 2017;16:1610-1620.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21:8787-8803.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Hetta HF. Gut immune response in the presence of hepatitis C virus infection. World J Immunol. 2014;4: 52.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121-141.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hetta HF, Mekky MA, Khalil NK, Mohamed WA, El-Feky MA, Ahmed SH, Daef EA, Medhat A, Nassar MI, Sherman KE, Shata MTM. Extra-hepatic infection of hepatitis C virus in the colon tissue and its relationship with hepatitis C virus pathogenesis. J Med Microbiol. 2016;65:703-712.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hetta HF, Mekky MA, Khalil NK, Mohamed WA, El-Feky MA, Ahmed SH, Daef EA, Nassar MI, Medhat A, Sherman KE, Shata MT. Association of colonic regulatory T cells with hepatitis C virus pathogenesis and liver pathology. J Gastroenterol Hepatol. 2015;30:1543-1551.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Mehta M, Hetta HF, Abdel-Hameed EA, Rouster SD, Hossain M, Mekky MA, Khalil NK, Mohamed WA, El-Feky MA, Ahmed SH, Daef EA, El-Mokhtar MA, Abdelwahab SF, Medhat A, Sherman KE, Shata MT. Association between IL28B rs12979860 single nucleotide polymorphism and the frequency of colonic Treg in chronically HCV-infected patients. Arch Virol. 2016;161:3161-3169.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology. 2014;146:1489-1499.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Conceição Neto N, Conceição-neto N, Zeller M, Lefrère H, De Bruyn P, Beller L, Deboutte W, Yinda CK, Lavigne R, Maes P, Van Ranst M, Matthijnssens J. NetoVIR: a reproducible protocol for virome analysis. Protoc Exch. 2016;.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Steen AD, Crits-Christoph A, Carini P, DeAngelis KM, Fierer N, Lloyd KG, Cameron Thrash J. High proportions of bacteria and archaea across most biomes remain uncultured. ISME J. 2019;13:3126-3130.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016;17:333-351.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Linde A. Single-field α-attractors. J Cosmol Astropart Phys. 2015;2015:003-003.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Zhao G, Vatanen T, Droit L, Park A, Kostic AD, Poon TW, Vlamakis H, Siljander H, Härkönen T, Hämäläinen AM, Peet A, Tillmann V, Ilonen J, Wang D, Knip M, Xavier RJ, Virgin HW. Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children. Proc Natl Acad Sci U S A. 2017;114:E6166-E6175.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Ma Y, You X, Mai G, Tokuyasu T, Liu C. A human gut phage catalog correlates the gut phageome with type 2 diabetes. Microbiome. 2018;6:24.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Coffey MJ, Low I, Stelzer-Braid S, Wemheuer B, Garg M, Thomas T, Jaffe A, Rawlinson WD, Ooi CY. The intestinal virome in children with cystic fibrosis differs from healthy controls. PLoS One. 2020;15:e0233557.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Bikel S, López-Leal G, Cornejo-Granados F, Gallardo-Becerra L, García-López R, Sánchez F, Equihua-Medina E, Ochoa-Romo JP, López-Contreras BE, Canizales-Quinteros S, Hernández-Reyna A, Mendoza-Vargas A, Ochoa-Leyva A. Gut dsDNA virome shows diversity and richness alterations associated with childhood obesity and metabolic syndrome. iScience. 2021;24:102900.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Yang K, Niu J, Zuo T, Sun Y, Xu Z, Tang W, Liu Q, Zhang J, Ng EKW, Wong SKH, Yeoh YK, Chan PKS, Chan FKL, Miao Y, Ng SC. Alterations in the Gut Virome in Obesity and Type 2 Diabetes Mellitus. Gastroenterology. 2021;161:1257-1269.e13.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Legoff J, Resche-Rigon M, Bouquet J, Robin M, Naccache SN, Mercier-Delarue S, Federman S, Samayoa E, Rousseau C, Piron P, Kapel N, Simon F, Socié G, Chiu CY. The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease. Nat Med. 2017;23:1080-1085.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Monaco CL, Gootenberg DB, Zhao G, Handley SA, Ghebremichael MS, Lim ES, Lankowski A, Baldridge MT, Wilen CB, Flagg M, Norman JM, Keller BC, Luévano JM, Wang D, Boum Y, Martin JN, Hunt PW, Bangsberg DR, Siedner MJ, Kwon DS, Virgin HW. Altered Virome and Bacterial Microbiome in Human Immunodeficiency Virus-Associated Acquired Immunodeficiency Syndrome. Cell Host Microbe. 2016;19:311-322.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Hannigan GD, Duhaime MB, Ruffin MT 4th, Koumpouras CC, Schloss PD. Diagnostic Potential and Interactive Dynamics of the Colorectal Cancer Virome. mBio. 2018;9.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Khan Mirzaei M, Khan MAA, Ghosh P, Taranu ZE, Taguer M, Ru J, Chowdhury R, Kabir MM, Deng L, Mondal D, Maurice CF. Bacteriophages Isolated from Stunted Children Can Regulate Gut Bacterial Communities in an Age-Specific Manner. Cell Host Microbe. 2020;27:199-212.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Jiang L, Lang S, Duan Y, Zhang X, Gao B, Chopyk J, Schwanemann LK, Ventura-Cots M, Bataller R, Bosques-Padilla F, Verna EC, Abraldes JG, Brown RS Jr, Vargas V, Altamirano J, Caballería J, Shawcross DL, Ho SB, Louvet A, Lucey MR, Mathurin P, Garcia-Tsao G, Kisseleva T, Brenner DA, Tu XM, Stärkel P, Pride D, Fouts DE, Schnabl B. Intestinal Virome in Patients With Alcoholic Hepatitis. Hepatology. 2020;72:2182-2196.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Zuo T, Zhan H, Zhang F, Liu Q, Tso EYK, Lui GCY, Chen N, Li A, Lu W, Chan FKL, Chan PKS, Ng SC. Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge. Gastroenterology. 2020;159:1302-1310.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Norman JM, Handley SA, Baldridge MT, Droit L, Liu CY, Keller BC, Kambal A, Monaco CL, Zhao G, Fleshner P, Stappenbeck TS, McGovern DP, Keshavarzian A, Mutlu EA, Sauk J, Gevers D, Xavier RJ, Wang D, Parkes M, Virgin HW. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell. 2015;160:447-460.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Berg G, Rybakova D, Fischer D, Cernava T, Vergès MC, Charles T, Chen X, Cocolin L, Eversole K, Corral GH, Kazou M, Kinkel L, Lange L, Lima N, Loy A, Macklin JA, Maguin E, Mauchline T, McClure R, Mitter B, Ryan M, Sarand I, Smidt H, Schelkle B, Roume H, Kiran GS, Selvin J, Souza RSC, van Overbeek L, Singh BK, Wagner M, Walsh A, Sessitsch A, Schloter M. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:103.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3:4-14.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Cai J, Sun L, Gonzalez FJ. Gut microbiota-derived bile acids in intestinal immunity, inflammation, and tumorigenesis. Cell Host Microbe. 2022;30:289-300.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Siddiqui MT, Cresci GAM. The Immunomodulatory Functions of Butyrate. J Inflamm Res. 2021;14:6025-6041.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Shkoporov AN, Hill C. Bacteriophages of the Human Gut: The "Known Unknown" of the Microbiome. Cell Host Microbe. 2019;25:195-209.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Carding SR, Davis N, Hoyles L. Review article: the human intestinal virome in health and disease. Aliment Pharmacol Ther. 2017;46:800-815.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Grove J, Marsh M. The cell biology of receptor-mediated virus entry. J Cell Biol. 2011;195:1071-1082.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Hetta HF, Rashed ZI, Ramadan YN, Al-Kadmy IMS, Kassem SM, Ata HS, Nageeb WM. Phage Therapy, a Salvage Treatment for Multidrug-Resistant Bacteria Causing Infective Endocarditis. Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Erez Z, Steinberger-Levy I, Shamir M, Doron S, Stokar-Avihail A, Peleg Y, Melamed S, Leavitt A, Savidor A, Albeck S, Amitai G, Sorek R. Communication between viruses guides lysis-lysogeny decisions. Nature. 2017;541:488-493.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Howard-Varona C, Hargreaves KR, Abedon ST, Sullivan MB. Lysogeny in nature: mechanisms, impact and ecology of temperate phages. ISME J. 2017;11:1511-1520.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Czyz A, Los M, Wrobel B, Wegrzyn G. Inhibition of spontaneous induction of lambdoid prophages in Escherichia coli cultures: simple procedures with possible biotechnological applications. BMC Biotechnol. 2001;1:1.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Obeng N, Pratama AA, Elsas JDV. The Significance of Mutualistic Phages for Bacterial Ecology and Evolution. Trends Microbiol. 2016;24:440-449.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Lopez CA, Winter SE, Rivera-Chávez F, Xavier MN, Poon V, Nuccio SP, Tsolis RM, Bäumler AJ. Phage-mediated acquisition of a type III secreted effector protein boosts growth of salmonella by nitrate respiration. mBio. 2012;3.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Jermyn WS, Boyd EF. Characterization of a novel Vibrio pathogenicity island (VPI-2) encoding neuraminidase (nanH) among toxigenic Vibrio cholerae isolates. Microbiology (Reading). 2002;148:3681-3693.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Brüssow H, Canchaya C, Hardt WD. Phages and the evolution of bacterial pathogens: from genomic rearrangements to lysogenic conversion. Microbiol Mol Biol Rev. 2004;68:560-602, table of contents.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Gödeke J, Paul K, Lassak J, Thormann KM. Phage-induced lysis enhances biofilm formation in Shewanella oneidensis MR-1. ISME J. 2011;5:613-626.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Wang X, Kim Y, Wood TK. Control and benefits of CP4-57 prophage excision in Escherichia coli biofilms. ISME J. 2009;3:1164-1179.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Lynch KH, Stothard P, Dennis JJ. Genomic analysis and relatedness of P2-like phages of the Burkholderia cepacia complex. BMC Genomics. 2010;11:599.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Abedon ST, Lejeune JT. Why Bacteriophage Encode Exotoxins and other Virulence Factors. Evol Bioinform Online. 2005;1:117693430500100.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Mauritzen JJ, Castillo D, Tan D, Svenningsen SL, Middelboe M. Beyond Cholera: Characterization of zot-Encoding Filamentous Phages in the Marine Fish Pathogen Vibrio anguillarum. Viruses. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Liang G, Bushman FD. The human virome: assembly, composition and host interactions. Nat Rev Microbiol. 2021;19:514-527.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Aggarwala V, Liang G, Bushman FD. Viral communities of the human gut: metagenomic analysis of composition and dynamics. Mob DNA. 2017;8:12.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Liang G, Zhao C, Zhang H, Mattei L, Sherrill-Mix S, Bittinger K, Kessler LR, Wu GD, Baldassano RN, DeRusso P, Ford E, Elovitz MA, Kelly MS, Patel MZ, Mazhani T, Gerber JS, Kelly A, Zemel BS, Bushman FD. The stepwise assembly of the neonatal virome is modulated by breastfeeding. Nature. 2020;581:470-474.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Sakata S, Tonooka T, Ishizeki S, Takada M, Sakamoto M, Fukuyama M, Benno Y. Culture-independent analysis of fecal microbiota in infants, with special reference to Bifidobacterium species. FEMS Microbiol Lett. 2005;243:417-423.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI. Human gut microbiome viewed across age and geography. Nature. 2012;486:222-227.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Manrique P, Bolduc B, Walk ST, van der Oost J, de Vos WM, Young MJ. Healthy human gut phageome. Proc Natl Acad Sci U S A. 2016;113:10400-10405.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Klaassens ES, de Vos WM, Vaughan EE. Metaproteomics approach to study the functionality of the microbiota in the human infant gastrointestinal tract. Appl Environ Microbiol. 2007;73:1388-1392.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Breitbart M, Haynes M, Kelley S, Angly F, Edwards RA, Felts B, Mahaffy JM, Mueller J, Nulton J, Rayhawk S, Rodriguez-Brito B, Salamon P, Rohwer F. Viral diversity and dynamics in an infant gut. Res Microbiol. 2008;159:367-373.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM, Warner BB, Tarr PI, Wang D, Holtz LR. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015;21:1228-1234.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Reyes A, Blanton LV, Cao S, Zhao G, Manary M, Trehan I, Smith MI, Wang D, Virgin HW, Rohwer F, Gordon JI. Gut DNA viromes of Malawian twins discordant for severe acute malnutrition. Proc Natl Acad Sci U S A. 2015;112:11941-11946.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Shkoporov AN, Clooney AG, Sutton TDS, Ryan FJ, Daly KM, Nolan JA, McDonnell SA, Khokhlova EV, Draper LA, Forde A, Guerin E, Velayudhan V, Ross RP, Hill C. The Human Gut Virome Is Highly Diverse, Stable, and Individual Specific. Cell Host Microbe. 2019;26:527-541.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Beller L, Deboutte W, Vieira-Silva S, Falony G, Tito RY, Rymenans L, Yinda CK, Vanmechelen B, Van Espen L, Jansen D, Shi C, Zeller M, Maes P, Faust K, Van Ranst M, Raes J, Matthijnssens J. The virota and its transkingdom interactions in the healthy infant gut. Proc Natl Acad Sci U S A. 2022;119:e2114619119.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Benler S, Yutin N, Antipov D, Raykov M, Shmakov S, Gussow AB, Pevzner PA, Koonin E. Thousands of previously unknown phages discovered in whole-community human gut metagenomes. Microbiome. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Gulyaeva A, Garmaeva S, Ruigrok RAAA, Wang D, Riksen NP, Netea MG, Wijmenga C, Weersma RK, Fu J, Vila AV, Kurilshikov A, Zhernakova A. Discovery, diversity, and functional associations of crAss-like phages in human gut metagenomes from four Dutch cohorts. Cell Rep. 2022;38:110204.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Dutilh BE, Cassman N, McNair K, Sanchez SE, Silva GG, Boling L, Barr JJ, Speth DR, Seguritan V, Aziz RK, Felts B, Dinsdale EA, Mokili JL, Edwards RA. A highly abundant bacteriophage discovered in the unknown sequences of human faecal metagenomes. Nat Commun. 2014;5:4498.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Espen LV, Bak EG, Beller L, Close L, Deboutte W, Juel HB, Nielsen T, Sinar D, Coninck LD, Frithioff-bøjsøe C, Fonvig CE, Jacobsen S, Kjærgaard M, Thiele M, Fullam A, Kuhn M, Holm J, Bork P, Krag A, Hansen T, Arumugam M, Matthijnssens J. A previously undescribed highly prevalent phage identified in a Danish enteric virome catalogue. mSystems. 2021;26:e0038221.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Camarillo-Guerrero LF, Almeida A, Rangel-Pineros G, Finn RD, Lawley TD. Massive expansion of human gut bacteriophage diversity. Cell. 2021;184:1098-1109.e9.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Devoto AE, Santini JM, Olm MR, Anantharaman K, Munk P, Tung J, Archie EA, Turnbaugh PJ, Seed KD, Blekhman R, Aarestrup FM, Thomas BC, Banfield JF. Megaphages infect Prevotella and variants are widespread in gut microbiomes. Nat Microbiol. 2019;4:693-700.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Krupovic M, Varsani A, Kazlauskas D, Breitbart M, Delwart E, Rosario K, Yutin N, Wolf YI, Harrach B, Zerbini FM, Dolja VV, Kuhn JH, Koonin EV. Cressdnaviricota: a Virus Phylum Unifying Seven Families of Rep-Encoding Viruses with Single-Stranded, Circular DNA Genomes. J Virol. 2020;94.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Zhang T, Breitbart M, Lee WH, Run JQ, Wei CL, Soh SW, Hibberd ML, Liu ET, Rohwer F, Ruan Y. RNA viral community in human feces: prevalence of plant pathogenic viruses. PLoS Biol. 2006;4:e3.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Schwarz D, Beuch U, Bandte M, Fakhro A, Büttner C, Obermeier C. Spread and interaction of Pepino mosaic virus (PepMV) and Pythium aphanidermatum in a closed nutrient solution recirculation system: effects on tomato growth and yield. Plant Pathology. 2010;59:443-452.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Reyes A, Haynes M, Hanson N, Angly FE, Heath AC, Rohwer F, Gordon JI. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature. 2010;466:334-338.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Garmaeva S, Gulyaeva A, Sinha T, Shkoporov AN, Clooney AG, Stockdale SR, Spreckels JE, Sutton TDS, Draper LA, Dutilh BE, Wijmenga C, Kurilshikov A, Fu J, Hill C, Zhernakova A. Stability of the human gut virome and effect of gluten-free diet. Cell Rep. 2021;35:109132.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Minot S, Bryson A, Chehoud C, Wu GD, Lewis JD, Bushman FD. Rapid evolution of the human gut virome. Proc Natl Acad Sci U S A. 2013;110:12450-12455.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Mosca A, Leclerc M, Hugot JP. Gut Microbiota Diversity and Human Diseases: Should We Reintroduce Key Predators in Our Ecosystem? Front Microbiol. 2016;7:455.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Matsuoka K, Kanai T. The gut microbiota and inflammatory bowel disease. Semin Immunopathol. 2015;37:47-55.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Lee M, Chang EB. Inflammatory Bowel Diseases (IBD) and the Microbiome-Searching the Crime Scene for Clues. Gastroenterology. 2021;160:524-537.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Carroll IM, Ringel-Kulka T, Siddle JP, Ringel Y. Alterations in composition and diversity of the intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil. 2012;24:521-530, e248.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Mayer EA, Savidge T, Shulman RJ. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology. 2014;146:1500-1512.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Pittayanon R, Lau JT, Yuan Y, Leontiadis GI, Tse F, Surette M, Moayyedi P. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. Gastroenterology. 2019;157:97-108.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, Goedert JJ, Hayes RB, Yang L. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 2013;105:1907-1911.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Yang J, Wei H, Zhou Y, Szeto CH, Li C, Lin Y, Coker OO, Lau HCH, Chan AWH, Sung JJY, Yu J. High-Fat Diet Promotes Colorectal Tumorigenesis Through Modulating Gut Microbiota and Metabolites. Gastroenterology. 2022;162:135-149.e2.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Saus E, Iraola-Guzmán S, Willis JR, Brunet-Vega A, Gabaldón T. Microbiome and colorectal cancer: Roles in carcinogenesis and clinical potential. Mol Aspects Med. 2019;69:93-106.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Moreno-Gallego JL, Chou SP, Di Rienzi SC, Goodrich JK, Spector TD, Bell JT, Youngblut ND, Hewson I, Reyes A, Ley RE. Virome Diversity Correlates with Intestinal Microbiome Diversity in Adult Monozygotic Twins. Cell Host Microbe. 2019;25:261-272.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Nishijima S, Nagata N, Kiguchi Y, Kojima Y, Miyoshi-Akiyama T, Kimura M, Ohsugi M, Ueki K, Oka S, Mizokami M, Itoi T, Kawai T, Uemura N, Hattori M. Extensive gut virome variation and its associations with host and environmental factors in a population-level cohort. Nat Commun. 2022;13:5252.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Gregory AC, Zablocki O, Zayed AA, Howell A, Bolduc B, Sullivan MB. The Gut Virome Database Reveals Age-Dependent Patterns of Virome Diversity in the Human Gut. Cell Host Microbe. 2020;28:724-740.e8.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Zuo T, Liu Q, Zhang F, Yeoh YK, Wan Y, Zhan H, Lui GC, Chen Z, Li AY, Cheung CP, Chen N, Lv W, Ng RW, Tso EY, Fung KS, Chan V, Ling L, Joynt G, Hui DS, Chan FK, Chan PK, Ng S. Temporal landscape of human gut RNA and DNA virome in SARS-CoV-2 infection and severity. Microbiome. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Mohr AE, Jäger R, Carpenter KC, Kerksick CM, Purpura M, Townsend JR, West NP, Black K, Gleeson M, Pyne DB, Wells SD, Arent SM, Kreider RB, Campbell BI, Bannock L, Scheiman J, Wissent CJ, Pane M, Kalman DS, Pugh JN, Ortega-Santos CP, Ter Haar JA, Arciero PJ, Antonio J. The athletic gut microbiota. J Int Soc Sports Nutr. 2020;17:24.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Zuo T, Sun Y, Wan Y, Yeoh YK, Zhang F, Cheung CP, Chen N, Luo J, Wang W, Sung JJY, Chan PKS, Wang K, Chan FKL, Miao Y, Ng SC. Human-Gut-DNA Virome Variations across Geography, Ethnicity, and Urbanization. Cell Host Microbe. 2020;28:741-751.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Minot S, Sinha R, Chen J, Li H, Keilbaugh SA, Wu GD, Lewis JD, Bushman FD. The human gut virome: inter-individual variation and dynamic response to diet. Genome Res. 2011;21:1616-1625.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Schulfer A, Santiago-Rodriguez TM, Ly M, Borin JM, Chopyk J, Blaser MJ, Pride DT. Fecal Viral Community Responses to High-Fat Diet in Mice. mSphere. 2020;5.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Lemay DG, Baldiviez LM, Chin EL, Spearman SS, Cervantes E, Woodhouse LR, Keim NL, Stephensen CB, Laugero KD. Technician-Scored Stool Consistency Spans the Full Range of the Bristol Scale in a Healthy US Population and Differs by Diet and Chronic Stress Load. J Nutr. 2021;151:1443-1452.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Aiemjoy K, Altan E, Aragie S, Fry DM, Phan TG, Deng X, Chanyalew M, Tadesse Z, Callahan EK, Delwart E, Keenan JD. Viral species richness and composition in young children with loose or watery stool in Ethiopia. BMC Infect Dis. 2019;19:53.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Maurice CF, Haiser HJ, Turnbaugh PJ. Xenobiotics shape the physiology and gene expression of the active human gut microbiome. Cell. 2013;152:39-50.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Sutcliffe SG, Shamash M, Hynes AP, Maurice CF. Common Oral Medications Lead to Prophage Induction in Bacterial Isolates from the Human Gut. Viruses. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Knowles B, Silveira CB, Bailey BA, Barott K, Cantu VA, Cobián-Güemes AG, Coutinho FH, Dinsdale EA, Felts B, Furby KA, George EE, Green KT, Gregoracci GB, Haas AF, Haggerty JM, Hester ER, Hisakawa N, Kelly LW, Lim YW, Little M, Luque A, McDole-Somera T, McNair K, de Oliveira LS, Quistad SD, Robinett NL, Sala E, Salamon P, Sanchez SE, Sandin S, Silva GG, Smith J, Sullivan C, Thompson C, Vermeij MJ, Youle M, Young C, Zgliczynski B, Brainard R, Edwards RA, Nulton J, Thompson F, Rohwer F. Lytic to temperate switching of viral communities. Nature. 2016;531:466-470.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Shamash M, Maurice CF. Phages in the infant gut: a framework for virome development during early life. ISME J. 2022;16:323-330.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Glassner KL, Abraham BP, Quigley EMM. The microbiome and inflammatory bowel disease. J Allergy Clin Immunol. 2020;145:16-27.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Friedrich M, Pohin M, Powrie F. Cytokine Networks in the Pathophysiology of Inflammatory Bowel Disease. Immunity. 2019;50:992-1006.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Guo X, Huang C, Xu J, Xu H, Liu L, Zhao H, Wang J, Huang W, Peng W, Chen Y, Nie Y, Zhou Y, Zhou Y. Gut Microbiota Is a Potential Biomarker in Inflammatory Bowel Disease. Front Nutr. 2021;8:818902.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Gasaly N, de Vos P, Hermoso MA. Impact of Bacterial Metabolites on Gut Barrier Function and Host Immunity: A Focus on Bacterial Metabolism and Its Relevance for Intestinal Inflammation. Front Immunol. 2021;12:658354.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17:223-237.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Khan I, Ullah N, Zha L, Bai Y, Khan A, Zhao T, Che T, Zhang C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Loddo I, Romano C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front Immunol. 2015;6:551.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Santos MPC, Gomes C, Torres J. Familial and ethnic risk in inflammatory bowel disease. Ann Gastroenterol. 2018;31:14-23.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Ruemmele FM. Role of Diet in Inflammatory Bowel Disease. Ann Nutr Metab. 2016;68 Suppl 1:33-41.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Zhang Z, Zhang H, Chen T, Shi L, Wang D, Tang D. Regulatory role of short-chain fatty acids in inflammatory bowel disease. Cell Commun Signal. 2022;20:64.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Jones DP, Richardson TG, Davey Smith G, Gunnell D, Munafò MR, Wootton RE. Exploring the Effects of Cigarette Smoking on Inflammatory Bowel Disease Using Mendelian Randomization. Crohns Colitis 360. 2020;2:otaa018.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut. 2005;54:1481-1491.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Birchenough G, Schroeder BO, Bäckhed F, Hansson GC. Dietary destabilisation of the balance between the microbiota and the colonic mucus barrier. Gut Microbes. 2019;10:246-250.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Clooney AG, Sutton TDS, Shkoporov AN, Holohan RK, Daly KM, O'Regan O, Ryan FJ, Draper LA, Plevy SE, Ross RP, Hill C. Whole-Virome Analysis Sheds Light on Viral Dark Matter in Inflammatory Bowel Disease. Cell Host Microbe. 2019;26:764-778.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Zuo T, Lu XJ, Zhang Y, Cheung CP, Lam S, Zhang F, Tang W, Ching JYL, Zhao R, Chan PKS, Sung JJY, Yu J, Chan FKL, Cao Q, Sheng JQ, Ng SC. Gut mucosal virome alterations in ulcerative colitis. Gut. 2019;68:1169-1179.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Pérez-Brocal V, García-López R, Nos P, Beltrán B, Moret I, Moya A. Metagenomic Analysis of Crohn's Disease Patients Identifies Changes in the Virome and Microbiome Related to Disease Status and Therapy, and Detects Potential Interactions and Biomarkers. Inflamm Bowel Dis. 2015;21:2515-2532.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Pérez-Brocal V, García-López R, Vázquez-Castellanos JF, Nos P, Beltrán B, Latorre A, Moya A. Study of the viral and microbial communities associated with Crohn's disease: a metagenomic approach. Clin Transl Gastroenterol. 2013;4:e36.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Lepage P, Colombet J, Marteau P, Sime-Ngando T, Doré J, Leclerc M. Dysbiosis in inflammatory bowel disease: a role for bacteriophages? Gut. 2008;57:424-425.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Wagner J, Maksimovic J, Farries G, Sim WH, Bishop RF, Cameron DJ, Catto-Smith AG, Kirkwood CD. Bacteriophages in gut samples from pediatric Crohn's disease patients: metagenomic analysis using 454 pyrosequencing. Inflamm Bowel Dis. 2013;19:1598-1608.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Wang W, Jovel J, Halloran B, Wine E, Patterson J, Ford G, OʼKeefe S, Meng B, Song D, Zhang Y, Tian Z, Wasilenko ST, Rahbari M, Reza S, Mitchell T, Jordan T, Carpenter E, Madsen K, Fedorak R, Dielemann LA, Ka-Shu Wong G, Mason AL. Metagenomic analysis of microbiome in colon tissue from subjects with inflammatory bowel diseases reveals interplay of viruses and bacteria. Inflamm Bowel Dis. 2015;21:1419-1427.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Fernandes MA, Verstraete SG, Phan TG, Deng X, Stekol E, LaMere B, Lynch SV, Heyman MB, Delwart E. Enteric Virome and Bacterial Microbiota in Children With Ulcerative Colitis and Crohn Disease. J Pediatr Gastroenterol Nutr. 2019;68:30-36.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Yan A, Butcher J, Mack D, Stintzi A. Virome Sequencing of the Human Intestinal Mucosal-Luminal Interface. Front Cell Infect Microbiol. 2020;10:582187.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Liang G, Conrad MA, Kelsen JR, Kessler LR, Breton J, Albenberg LG, Marakos S, Galgano A, Devas N, Erlichman J, Zhang H, Mattei L, Bittinger K, Baldassano RN, Bushman FD. Dynamics of the Stool Virome in Very Early-Onset Inflammatory Bowel Disease. J Crohns Colitis. 2020;14:1600-1610.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Nishiyama H, Endo H, Blanc-Mathieu R, Ogata H. Ecological Structuring of Temperate Bacteriophages in the Inflammatory Bowel Disease-Affected Gut. Microorganisms. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Imai T, Inoue R, Nishida A, Yokota Y, Morishima S, Kawahara M, Kusada H, Tamaki H, Andoh A. Features of the gut prokaryotic virome of Japanese patients with Crohn's disease. J Gastroenterol. 2022;57:559-570.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Duerkop BA, Kleiner M, Paez-Espino D, Zhu W, Bushnell B, Hassell B, Winter SE, Kyrpides NC, Hooper LV. Murine colitis reveals a disease-associated bacteriophage community. Nat Microbiol. 2018;3:1023-1031.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Reyes A, Wu M, McNulty NP, Rohwer FL, Gordon JI. Gnotobiotic mouse model of phage-bacterial host dynamics in the human gut. Proc Natl Acad Sci U S A. 2013;110:20236-20241.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, Casero D, Courtney H, Gonzalez A, Graeber TG, Hall AB, Lake K, Landers CJ, Mallick H, Plichta DR, Prasad M, Rahnavard G, Sauk J, Shungin D, Vázquez-Baeza Y, White RA 3rd; IBDMDB Investigators, Braun J, Denson LA, Jansson JK, Knight R, Kugathasan S, McGovern DPB, Petrosino JF, Stappenbeck TS, Winter HS, Clish CB, Franzosa EA, Vlamakis H, Xavier RJ, Huttenhower C. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569:655-662.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, Chandra J, Retuerto MA, Neut C, Vermeire S, Clemente J, Colombel JF, Fujioka H, Poulain D, Sendid B, Ghannoum MA. Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn's Disease. mBio. 2016;7.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780-13785.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Uzbay T. Germ-free animal experiments in the gut microbiota studies. Curr Opin Pharmacol. 2019;49:6-10.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Qv L, Yang Z, Yao M, Mao S, Li Y, Zhang J, Li L. Methods for Establishment and Maintenance of Germ-Free Rat Models. Front Microbiol. 2020;11:1148.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Hsu BB, Gibson TE, Yeliseyev V, Liu Q, Bry L, Silver PA, Gerber GK. Bacteriophages dynamically modulate the gut microbiota and metabolome. Cell Host Microbe. 2019;25:803-814.e5.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Sinha A, Li Y, Mirzaei MK, Shamash M, Samadfam R, King IL, Maurice CF. Transplantation of bacteriophages from ulcerative colitis patients shifts the gut bacteriome and exacerbates the severity of DSS colitis. Microbiome. 2022;10:105.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Lin DM, Lin HC. A theoretical model of temperate phages as mediators of gut microbiome dysbiosis. F1000Res. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Abeles SR, Pride DT. Molecular bases and role of viruses in the human microbiome. J Mol Biol. 2014;426:3892-3906.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Babickova J, Gardlik R. Pathological and therapeutic interactions between bacteriophages, microbes and the host in inflammatory bowel disease. World J Gastroenterol. 2015;21:11321-11330.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Cornuault JK, Petit MA, Mariadassou M, Benevides L, Moncaut E, Langella P, Sokol H, De Paepe M. Phages infecting Faecalibacterium prausnitzii belong to novel viral genera that help to decipher intestinal viromes. Microbiome. 2018;6:65.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Zuppi M, Hendrickson HL, O'Sullivan JM, Vatanen T. Phages in the Gut Ecosystem. Front Cell Infect Microbiol. 2021;11:822562.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805-820.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  El-zayat SR, Sibaii H, Mannaa FA. Toll-like receptors activation, signaling, and targeting: an overview. Bull Natl Res Cent. 2019;43:187.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Metzger RN, Krug AB, Eisenächer K. Enteric Virome Sensing-Its Role in Intestinal Homeostasis and Immunity. Viruses. 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Virgin HW. The virome in mammalian physiology and disease. Cell. 2014;157:142-150.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Rokutan K, Kawahara T, Kuwano Y, Tominaga K, Nishida K, Teshima-Kondo S. Nox enzymes and oxidative stress in the immunopathology of the gastrointestinal tract. Semin Immunopathol. 2008;30:315-327.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Lu Y, Li X, Liu S, Zhang Y, Zhang D. Toll-like Receptors and Inflammatory Bowel Disease. Front Immunol. 2018;9:72.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Ihara S, Hirata Y, Koike K. TGF-β in inflammatory bowel disease: a key regulator of immune cells, epithelium, and the intestinal microbiota. J Gastroenterol. 2017;52:777-787.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Atri C, Guerfali FZ, Laouini D. Role of Human Macrophage Polarization in Inflammation during Infectious Diseases. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Gogokhia L, Buhrke K, Bell R, Hoffman B, Brown DG, Hanke-Gogokhia C, Ajami NJ, Wong MC, Ghazaryan A, Valentine JF, Porter N, Martens E, O'Connell R, Jacob V, Scherl E, Crawford C, Stephens WZ, Casjens SR, Longman RS, Round JL. Expansion of Bacteriophages Is Linked to Aggravated Intestinal Inflammation and Colitis. Cell Host Microbe. 2019;25:285-299.e8.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Gogokhia L, Round JL. Immune-bacteriophage interactions in inflammatory bowel diseases. Curr Opin Virol. 2021;49:30-35.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Barr JJ, Auro R, Furlan M, Whiteson KL, Erb ML, Pogliano J, Stotland A, Wolkowicz R, Cutting AS, Doran KS, Salamon P, Youle M, Rohwer F. Bacteriophage adhering to mucus provide a non-host-derived immunity. Proc Natl Acad Sci U S A. 2013;110:10771-10776.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Kernbauer E, Ding Y, Cadwell K. An enteric virus can replace the beneficial function of commensal bacteria. Nature. 2014;516:94-98.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Rahier JF, Magro F, Abreu C, Armuzzi A, Ben-Horin S, Chowers Y, Cottone M, de Ridder L, Doherty G, Ehehalt R, Esteve M, Katsanos K, Lees CW, Macmahon E, Moreels T, Reinisch W, Tilg H, Tremblay L, Veereman-Wauters G, Viget N, Yazdanpanah Y, Eliakim R, Colombel JF; European Crohn's and Colitis Organisation (ECCO). Second European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease. J Crohns Colitis. 2014;8:443-468.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Ungaro F, Massimino L, Furfaro F, Rimoldi V, Peyrin-Biroulet L, D'Alessio S, Danese S. Metagenomic analysis of intestinal mucosa revealed a specific eukaryotic gut virome signature in early-diagnosed inflammatory bowel disease. Gut Microbes. 2019;10:149-158.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Santiago-Rodriguez TM, Hollister EB. Human Virome and Disease: High-Throughput Sequencing for Virus Discovery, Identification of Phage-Bacteria Dysbiosis and Development of Therapeutic Approaches with Emphasis on the Human Gut. Viruses. 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Scarpellini E, Ianiro G, Attili F, Bassanelli C, De Santis A, Gasbarrini A. The human gut microbiota and virome: Potential therapeutic implications. Dig Liver Dis. 2015;47:1007-1012.  [PubMed]  [DOI]  [Cited in This Article: ]
154.  Beller L, Matthijnssens J. What is (not) known about the dynamics of the human gut virome in health and disease. Curr Opin Virol. 2019;37:52-57.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Basic M, Keubler LM, Buettner M, Achard M, Breves G, Schröder B, Smoczek A, Jörns A, Wedekind D, Zschemisch NH, Günther C, Neumann D, Lienenklaus S, Weiss S, Hornef MW, Mähler M, Bleich A. Norovirus triggered microbiota-driven mucosal inflammation in interleukin 10-deficient mice. Inflamm Bowel Dis. 2014;20:431-443.  [PubMed]  [DOI]  [Cited in This Article: ]
156.  Cadwell K, Patel KK, Maloney NS, Liu TC, Ng AC, Storer CE, Head RD, Xavier R, Stappenbeck TS, Virgin HW. Virus-plus-susceptibility gene interaction determines Crohn's disease gene Atg16L1 phenotypes in intestine. Cell. 2010;141:1135-1145.  [PubMed]  [DOI]  [Cited in This Article: ]
157.  Massimino L, Palmieri O, Facoetti A, Fuggetta D, Spanò S, Lamparelli LA, D'Alessio S, Cagliani S, Furfaro F, D'Amico F, Zilli A, Fiorino G, Parigi TL, Noviello D, Latiano A, Bossa F, Latiano T, Pirola A, Mologni L, Piazza RG, Abbati D, Perri F, Bonini C, Peyrin-Biroulet L, Malesci A, Jairath V, Danese S, Ungaro F. Gut virome-colonising Orthohepadnavirus genus is associated with ulcerative colitis pathogenesis and induces intestinal inflammation in vivo. Gut. 2023;72:1838-1847.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Dave M, Higgins PD, Middha S, Rioux KP. The human gut microbiome: current knowledge, challenges, and future directions. Transl Res. 2012;160:246-257.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Dutilh BE, Reyes A, Hall RJ, Whiteson KL. Editorial: Virus Discovery by Metagenomics: The (Im)possibilities. Front Microbiol. 2017;8:1710.  [PubMed]  [DOI]  [Cited in This Article: ]
160.  Zhang YZ, Shi M, Holmes EC. Using Metagenomics to Characterize an Expanding Virosphere. Cell. 2018;172:1168-1172.  [PubMed]  [DOI]  [Cited in This Article: ]
161.  Kleiner M, Hooper LV, Duerkop BA. Evaluation of methods to purify virus-like particles for metagenomic sequencing of intestinal viromes. BMC Genomics. 2015;16:7.  [PubMed]  [DOI]  [Cited in This Article: ]
162.  Nayfach S, Camargo AP, Schulz F, Eloe-Fadrosh E, Roux S, Kyrpides NC. CheckV assesses the quality and completeness of metagenome-assembled viral genomes. Nat Biotechnol. 2021;39:578-585.  [PubMed]  [DOI]  [Cited in This Article: ]
163.  Gloor GB, Hummelen R, Macklaim JM, Dickson RJ, Fernandes AD, MacPhee R, Reid G. Microbiome profiling by illumina sequencing of combinatorial sequence-tagged PCR products. PLoS One. 2010;5:e15406.  [PubMed]  [DOI]  [Cited in This Article: ]
164.  Ren J, Ahlgren NA, Lu YY, Fuhrman JA, Sun F. VirFinder: a novel k-mer based tool for identifying viral sequences from assembled metagenomic data. Microbiome. 2017;5:69.  [PubMed]  [DOI]  [Cited in This Article: ]
165.  Li Y, Wang H, Nie K, Zhang C, Zhang Y, Wang J, Niu P, Ma X. VIP: an integrated pipeline for metagenomics of virus identification and discovery. Sci Rep. 2016;6:23774.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Roux S, Tournayre J, Mahul A, Debroas D, Enault F. Metavir 2: new tools for viral metagenome comparison and assembled virome analysis. BMC Bioinformatics. 2014;15:76.  [PubMed]  [DOI]  [Cited in This Article: ]
167.  Metsky HC, Siddle KJ, Gladden-Young A, Qu J, Yang DK, Brehio P, Goldfarb A, Piantadosi A, Wohl S, Carter A, Lin AE, Barnes KG, Tully DC, Corleis B, Hennigan S, Barbosa-Lima G, Vieira YR, Paul LM, Tan AL, Garcia KF, Parham LA, Odia I, Eromon P, Folarin OA, Goba A; Viral Hemorrhagic Fever Consortium, Simon-Lorière E, Hensley L, Balmaseda A, Harris E, Kwon DS, Allen TM, Runstadler JA, Smole S, Bozza FA, Souza TML, Isern S, Michael SF, Lorenzana I, Gehrke L, Bosch I, Ebel G, Grant DS, Happi CT, Park DJ, Gnirke A, Sabeti PC, Matranga CB. Capturing sequence diversity in metagenomes with comprehensive and scalable probe design. Nat Biotechnol. 2019;37:160-168.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Costea PI, Hildebrand F, Arumugam M, Bäckhed F, Blaser MJ, Bushman FD, de Vos WM, Ehrlich SD, Fraser CM, Hattori M, Huttenhower C, Jeffery IB, Knights D, Lewis JD, Ley RE, Ochman H, O'Toole PW, Quince C, Relman DA, Shanahan F, Sunagawa S, Wang J, Weinstock GM, Wu GD, Zeller G, Zhao L, Raes J, Knight R, Bork P. Enterotypes in the landscape of gut microbial community composition. Nat Microbiol. 2018;3:8-16.  [PubMed]  [DOI]  [Cited in This Article: ]
169.  Holmes I, Harris K, Quince C. Dirichlet multinomial mixtures: generative models for microbial metagenomics. PLoS One. 2012;7:e30126.  [PubMed]  [DOI]  [Cited in This Article: ]
170.  Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis JD. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105-108.  [PubMed]  [DOI]  [Cited in This Article: ]
171.  Vieira-Silva S, Falony G, Belda E, Nielsen T, Aron-Wisnewsky J, Chakaroun R, Forslund SK, Assmann K, Valles-Colomer M, Nguyen TTD, Proost S, Prifti E, Tremaroli V, Pons N, Le Chatelier E, Andreelli F, Bastard JP, Coelho LP, Galleron N, Hansen TH, Hulot JS, Lewinter C, Pedersen HK, Quinquis B, Rouault C, Roume H, Salem JE, Søndertoft NB, Touch S; MetaCardis Consortium, Dumas ME, Ehrlich SD, Galan P, Gøtze JP, Hansen T, Holst JJ, Køber L, Letunic I, Nielsen J, Oppert JM, Stumvoll M, Vestergaard H, Zucker JD, Bork P, Pedersen O, Bäckhed F, Clément K, Raes J. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature. 2020;581:310-315.  [PubMed]  [DOI]  [Cited in This Article: ]
172.  Vandeputte D, Kathagen G, D'hoe K, Vieira-Silva S, Valles-Colomer M, Sabino J, Wang J, Tito RY, De Commer L, Darzi Y, Vermeire S, Falony G, Raes J. Quantitative microbiome profiling links gut community variation to microbial load. Nature. 2017;551:507-511.  [PubMed]  [DOI]  [Cited in This Article: ]
173.  Vieira-Silva S, Sabino J, Valles-Colomer M, Falony G, Kathagen G, Caenepeel C, Cleynen I, van der Merwe S, Vermeire S, Raes J. Quantitative microbiome profiling disentangles inflammation- and bile duct obstruction-associated microbiota alterations across PSC/IBD diagnoses. Nat Microbiol. 2019;4:1826-1831.  [PubMed]  [DOI]  [Cited in This Article: ]
174.  Valles-Colomer M, Bacigalupe R, Vieira-Silva S, Suzuki S, Darzi Y, Tito RY, Yamada T, Segata N, Raes J, Falony G. Variation and transmission of the human gut microbiota across multiple familial generations. Nat Microbiol. 2022;7:87-96.  [PubMed]  [DOI]  [Cited in This Article: ]
175.  Fromentin S, Forslund SK, Chechi K, Aron-Wisnewsky J, Chakaroun R, Nielsen T, Tremaroli V, Ji B, Prifti E, Myridakis A, Chilloux J, Andrikopoulos P, Fan Y, Olanipekun MT, Alves R, Adiouch S, Bar N, Talmor-Barkan Y, Belda E, Caesar R, Coelho LP, Falony G, Fellahi S, Galan P, Galleron N, Helft G, Hoyles L, Isnard R, Le Chatelier E, Julienne H, Olsson L, Pedersen HK, Pons N, Quinquis B, Rouault C, Roume H, Salem JE, Schmidt TSB, Vieira-Silva S, Li P, Zimmermann-Kogadeeva M, Lewinter C, Søndertoft NB, Hansen TH, Gauguier D, Gøtze JP, Køber L, Kornowski R, Vestergaard H, Hansen T, Zucker JD, Hercberg S, Letunic I, Bäckhed F, Oppert JM, Nielsen J, Raes J, Bork P, Stumvoll M, Segal E, Clément K, Dumas ME, Ehrlich SD, Pedersen O. Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat Med. 2022;28:303-314.  [PubMed]  [DOI]  [Cited in This Article: ]
176.  Song L, Zhang L, Fang X. Characterizing enterotypes in human metagenomics: a viral perspective. Front. Microbiol.12:740990.  [PubMed]  [DOI]  [Cited in This Article: ]
177.  Jansen D, Falony G, Vieira-Silva S, Simsek C, Marcelis T, Caenepeel C, Machiels K, Raes J, Vermeire S, Matthijnssens J. Community Types of the Human Gut Virome are Associated with Endoscopic Outcome in Ulcerative Colitis. J Crohns Colitis. 2023;17:1504-1513.  [PubMed]  [DOI]  [Cited in This Article: ]
178.  Qv L, Mao S, Li Y, Zhang J, Li L. Roles of Gut Bacteriophages in the Pathogenesis and Treatment of Inflammatory Bowel Disease. Front Cell Infect Microbiol. 2021;11:755650.  [PubMed]  [DOI]  [Cited in This Article: ]