Next Article in Journal
Pediatric Bone Marrow Failure: A Broad Landscape in Need of Personalized Management
Previous Article in Journal
Cardiac Magnetic Resonance Identifies Responders to Cardiac Resynchronization Therapy with an Assessment of Septal Scar and Left Ventricular Dyssynchrony
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evolution of a Pathogenic Microbiome

by
Pui Yin Wong
1,†,
Carmen Yip
1,†,
Daniel A. Lemberg
2,
Andrew S. Day
1,2,3 and
Steven T. Leach
1,*
1
Discipline of Paediatrics, School of Clinical Medicine, University of NSW, Sydney 2052, Australia
2
Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia
3
Department of Paediatrics, University of Otago, Christchurch 8011, New Zealand
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2023, 12(22), 7184; https://doi.org/10.3390/jcm12227184
Submission received: 22 September 2023 / Revised: 16 November 2023 / Accepted: 16 November 2023 / Published: 20 November 2023

Abstract

:
The process of microbiome development arguably begins before birth. Vertical transmission of bacteria from the mother to the infant is a keystone event in microbiome development. Subsequent to birth, the developing microbiome is vulnerable to influence from a wide range of factors. Additionally, the microbiome can influence the health and development of the host infant. This intricate interaction of the gastrointestinal microbiome and the host has been described as both symbiotic and dysbiotic. Defining these terms, a symbiotic microbiome is where the microbiome and host provide mutual benefit to each other. A pathogenic microbiome, or more precisely a gastrointestinal microbiome associated with disease, is increasing described as dysbiotic. This review seeks to investigate the factors that contribute to evolving a disease-causing or ‘dysbiotic’ microbiome. This review covers the development of the gastrointestinal microbiome in infants, the interaction of the microbiome with the host, and its contribution to host immunity and investigates specific features of the gastrointestinal microbiome that are associated with disease.

Graphical Abstract

1. Introduction

There are estimated to be approximately one hundred trillion microbes in or on the body, which outnumber human cells at a ratio of 1.3:l [1]. Most of these microbes are located in the gut to form the gastrointestinal (GI) microbiome [2]. This microbiome consists of bacteria, viruses, archaea, and eukaryotes from over 1500 species, contributing to various physiological functions [3]. This microbiome has integral roles in human health, immune development, and nutrient metabolism [4,5,6]. However, alterations in this microbiome can contribute to inflammation and have also been closely associated with the occurrence of multiple diseases [2].
The origin of the gut microbiome is multi-faceted. The very first exposure being the maternal microbiome, which is vertically transmitted during the perinatal period and determines the first colonisers in the infant [7]. Thus, mother-to-infant transfer is an important checkpoint in early life, defining the acquisition and succession of the early-life microbiome [8,9]. Emerging evidence suggests that this process occurs in various patterns, durations, and body sites. It is affected by a multitude of factors, all with the capacity to alter the infant’s microbiome [10,11,12,13]. The recent recognition that the modulation of the early gut microbiome offers great potential to exert substantial changes in health outcomes has attracted great interest towards this field. This review aims to provide an overview of the evolution of the GI microbiome and explores the concept of a pathogenic microbiome.

2. Vertical Transmission of the Maternal Microbiome

2.1. Transmission of the Placental Microbiome

The placenta is an organ that plays a vital role in the development of the foetus, responsible for the frequent exchange of nutrients, oxygen, and waste products between the mother and foetus [14]. Traditionally, the placenta has been thought of as a sterile environment free of microorganisms. One of the earliest studies supporting the sterility of the placenta, which utilised 16S rRNA sequencing, provided evidence that there was an indistinguishable difference of bacterial species copy numbers between placental biopsies and study controls [15]. This was subsequently corroborated by Lager et al. [16] who utilised 18S rRNA sequencing and detected no eukaryotic signals in placental samples of women with adverse pregnancy outcomes and healthy controls. Several studies also attributed the putative existence of a prenatal microbiome to the contamination of kits and reagents in the laboratory [15,17,18].
However, over the past decade, emerging studies have contradicted the dogma of a sterile placenta. The vertical transmission of bacteria via the placenta is thought to occur through two main routes—haematogenous and ascendant [19]. The former occurs where the microorganisms cross the placenta and colonise the foetal gut, whereas the latter refers to microorganisms resident in the cervix, vagina, or other body sites ascending to the uterus to colonise the foetal gut [19]. In 2020 and 2021, two studies conducted by Rackaityte et al. [20] and Mishra et al. [21], respectively, provided more convincing evidence to support this hypothesis. Microbial signals were consistently detected across different foetal organs including the placenta [20]. Specifically, Lactobacillus and Staphylococcus were identified as the most prevalent [20]. Experiments conducted in vitro confirmed that bacterial antigens in utero induced the activation of memory T-cells in foetal mesenteric lymph nodes, as opposed to contamination from the external environment [21]. These findings allude to the existence of viable microbes in utero and the extent to which a low-biomass community can promote foetal development before birth. Nonetheless, the mechanisms of vertical transmission through the placenta are not yet fully understood and the contamination in low-biomass samples argues against this possibility. Further studies with rigorous aseptic techniques and novel technologies to improve the sensitivity in the analysis of low-biomass samples is needed to understand the importance of placental microbiome transfer.

2.2. Transmission of the Vaginal Microbiome

The mode of delivery is accepted as having a crucial role in early gut microbiome colonisation. During vaginal delivery, the neonate is first exposed to the vaginal and gut microbiome of the mother [10]. The major microbiome that predominates the infant’s gut is hence the maternal vaginal microbiome, with a minor component being the external environment [10,22]. Studies have shown that the maternal vaginal microbiome constitutes up to 16.3% of the infant’s total gut microbiome at day 1, which is not observed in infants delivered by Caesarean section [8].
Several studies have investigated the transmission of the microbiome during delivery, with the consensus that the microbial community of vaginally delivered neonates exhibit a diverse microbiome that is constituted from maternal vaginal and intestinal bacteria [10,23,24]. This is dominated by Lactobacillus, Prevotella, Bifidobacterium, Parabacteroides, and Escherichia [10,23,24]. Conversely, infants delivered via Caesarean section are found to have increased relative abundances of Klebsiella, Clostridia, Enterobacter, Staphylococcus, and opportunistic pathogens, mainly acquired from the maternal skin and hospital environment [8,25,26].
Bifidobacterium and Lactobacillus are essential in promoting immunogenic humoral and cell-mediated maturation, with immediate roles in the development and protection of the gut microbiome against pathogenic bacteria [27]. However, in Caesarean-delivered infants there can be delays of up to 4 months to acquire key bacteria [26].
The persistence patterns of bacteria in vaginally delivered infants also vary. These infants are enriched with Lactobacillus, Bifidobacterium bifidum, Bifidobacterium longum, and Parabacteroides diastonis, and these bacteria are better colonisers as opposed to Streptococcus salivarius, Staphylococcus massiliensis, Staphylococcus hominis, and Veillonella parvula, which tend to only be in the gut transiently [8]. A possible explanation is that vertically transferred bacteria are better adapted to the intestinal environment and therefore have a competitive advantage in persisting in the infant’s microbiome.

2.3. Transmission of Breast Milk Microbiome

Breast milk accounts for the second most common source of microorganisms after the birth canal for infants that are born vaginally [28]. Studies have proposed that microbes translocate from the maternal gut to the mammary gland, with some influences from the maternal skin and oral cavity during breastfeeding [29,30,31].
The World Health Organisation (WHO) recommends exclusive breastfeeding for at least the first six months of life, following which an introduction to solid foods should commence [32]. It is estimated that 1–6-month-old infants, with an average breastmilk consumption of 670 mL/day [33], consume up to 700,000 bacteria daily, serving as a substantial coloniser of the infant gut [34]. Breast milk possesses a complex and dynamic composition that fulfils an infant’s nutritional requirements and contains certain bioactive components including human milk oligosaccharides (HMOs), antibodies, immune cells, antimicrobial proteins, cytokines, peptides, and lactoferrin [35]. These largely help strengthen the immune system of the newborn and offer protection against disease [35]. Previous research showed that non-breastfed infants are at a substantially increased risk of GI and respiratory infections, atopy, and chronic conditions, substantiating the antimicrobial and immunoregulatory properties of breastmilk and potentially breastmilk-derived bacteria [5,36,37].
It is clear that breastmilk contributes to a significant proportion of an infant’s gut microbiome in the first year of life [38,39,40,41]. Utilising strain-level analyses and metagenomic sequencing, several studies identified shared bacterial strains between maternal milk or faecal samples, and infant faecal samples [38,39,40,41]. Several bacterial strains were consistently identified, with the most prominent being Bifidobacterium breve and B. longum [38,39,40,41]. Although the sample sizes of these studies were small and negative controls were not included in all the studies, the consistency of the results support associations between breast milk and the development of the infant gut microbiome.
A larger randomised control trial with 107 mother–infant dyads found that the breast milk bacterial communities were significantly associated with the infant gut microbiome during the first 4 months of life [28]. Bifidobacterium, Lactobacilli, and Staphylococci, which dominated the milk of lactating mothers, were observed at an increased abundance and diversity in the stool samples of breastfed infants compared to non-breastfed infants [28]. The number of shared microbes and bacterial diversity also increased with the proportion of breastmilk intake in a dose-dependent manner [28]. Furthermore, each infant’s gut microbiome resistome was found to be more similar to their respective mother’s microbiome than that of an unrelated woman [42,43].
This evidence suggests that breastmilk supports the growth of organisms in the infant’s gut, and acts as a potential site of microbial transmission between mothers and infants. However, data surrounding the proportion of shared microbes should be interpreted with caution, as it remains unclear whether the bacterial communities identified in the infant stool samples are derived from sources other than breastmilk. A primary limitation of these studies was the taxonomic resolution, which limited identification of the bacteria to the genus or species level; these genus or species are commonly found at various body sites and across different individuals. Furthermore, most of the studies investigating the vertical transmission of breast milk also contained limited numbers of dyad pairs and limited culturing of targeted microbes. As such, further studies with larger sample sizes and strain-level analysis will assist in overcoming previous limitations and provide more insight into the role of breast milk in the development of the infant’s microbiome.

2.4. Other Potential Sources of Transmission

The other potential sources of vertical transmission include the maternal oral cavity and skin. The relative abundance of shared species in the oral cavity and skin accounted for approximately 7% and 5% of the infant’s microbiome, respectively [8]. They were reported to be more pronounced for up to the first three days of life but decreased with time, suggesting that they are transient colonisers of the gut [8]. Though these sites are not significant contributors to the infant’s initial bacterial community, they may have an influence in later life, opening an avenue for further research.

3. Development of the Gut Microbiome from 0 to 6 Months

Within 24 h of birth there is a high diversity of species observed in the infant faecal microbiome, reflecting the rapid influx of pioneering microbes from multiple sources. Diversity and richness then rapidly decrease as unsuitable or poorly adapted organisms are lost or replaced [8,44]. Common species observed at day 1 but not later include: Clostridium innocuum, Streptococcus parasanguinis, Prevotella melaningonica, Alistipes putredinis, and Haemophilus parainfluenzae [44]. This pattern suggests an influx of species stochastically, followed by the establishment of a core set of species that persists stably in the gut. Of interest, Chu et al. [23] studied 117 stool samples and identified Klebsiella and Escherichia as the genera with the highest specificity to the neonate’s gut. Notably, these taxa are known facultative anaerobes that are typically observed in the early GI tract, as the newborn gut is predominantly an aerobic environment at birth.
In the weeks following birth, the GI environment becomes anaerobic, thereby favouring the colonisation of strict anaerobes. This is observed by the prevailing taxa in the infant’s gut changing from mostly facultative anaerobes to strict anaerobes—namely Bifidobacterium, Clostridium, and Bacteroides [38,45,46]. At this stage, when the primary source of an infant’s nutrition is breastmilk, the infant’s gut favours the propagation of certain microbes such as Bifidobacterium that selectively metabolise HMO in breastmilk. This provides Bifidobacterium a nutrient source unavailable to other colonisers including potential pathogens, allowing it to thrive in the developing microbiome [9,47]. Bifidobacterium and HMO act synergistically to provide optimal protection to the infant due to their immunogenic roles in preventing the attachment of pathogens, suppressing inflammation, and promoting immune development [47]. In a healthy, vaginally delivered, full-term and breastfed infant, Bifidobacterium is the dominant bacteria 1–2 weeks after birth. However, Bifidobacterium is then reduced at 4–6 months of age with weaning and after the addition of solid foods to the infant diet as other environmental factors continue to alter the gut microbiome [8,45].
With the introduction of solid food and cessation of breastfeeding there are increased abundances of Bacteroides and Clostridiales [8,45]. This is associated with an increase in the functional capacity of the microbiome specifically in its capacity to degrade complex sugars, starch, and increase vitamin biosynthesis [8,45]. The explanation for this change is attributed to infant diet modification, as this new diet consists of many polysaccharides, which their immature digestive system cannot digest, thereby trigging an increase in these microbes to facilitate this process [48]. From this point, the alpha diversity and composition of the infant gut microbiome continues maturing until five years of age, and steadily begins to resemble the adult-like structure [8,49].
It is also important to note that different geographical locations exhibit variations in gut microbial composition. In a study by Kuang et al. [50] Proteobacteria, particularly Enterobacteriaceae, were found to be the dominant type of bacteria in Chinese infants <3 months of age. In contrast, Actinobacteria, especially Bifidobacterium, were found to be the dominant genera of the intestinal microbiome of Western infants, with a smaller bacterial diversity compared to infants in developing countries [50]. Moreover, Enterobacteria were present in Pakistani and Chinese infants during the first weeks of life while they were only detected in Swedish infants at 6 months of age [51,52].
The variability in the rate of acquisition and gut microbiome profiles are hypothesised to be driven by many factors including host genetics [53], sanitation levels [52], variations of vaginal and skin microbial composition in mothers [52], and higher formula feeding rates in Western and other upper-middle income countries [54]. It is thus evident that regional variances contribute to the interindividual compositional differences of an infant’s microbiome. Nevertheless, it is accepted that by around the age of five years, the infant microbiome has essentially developed and fulfills the functions of an ‘adult’ microbiome.

4. Composition and Function of the Developed Microbiome

In a healthy adult, the gut microbiome is predominated by the phyla Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucomicrobia [55,56]. The adult intestinal microbiome also contains a gene set approximately 150 times larger than that of the human genome [55,57]. Through symbiotic interactions with host structures, this microbiome participates in metabolic and immunological processes such as the digestion of nutrients and drugs, synthesis of vitamins, and immunomodulation to maintain homeostasis [3,58,59,60].
However, the specific composition of the gut microbiome varies widely between healthy individuals [61]. Therefore, to understand the gut microbiome requires more than defining set quantities of specific species of microbes, but requires an understanding of the microbiome functional capacity [61,62]. Overall, a healthy microbiome exhibits diversity, stability, and an ability to fulfil its metabolic activities [63,64]. The richer and more diverse the microbiome, the more resilient it is to external threats, providing an inertia to maintain equilibrium [55,61]. A healthy and functional microbiome also promotes competitive exclusion, preventing the overgrowth of pathogenic bacteria [58].

5. The Microbiome and Host Immunity

The microbiome plays an integral role in host immunity. The initial line of defense in host immunity is the intestinal epithelial barrier, which protects the host via a mucous layer against luminal microbes [65]. The microbiome can assist the host in maintaining tolerance to beneficial commensal bacteria by preventing the overgrowth of pathogenic strains [2]. The microbiome can also induce the host to synthesise antimicrobial proteins [3,59] and express local immunoglobulins, specifically secretory immunoglobulin A (IgA) [58].
The microbiome also aids the maturation of the innate and adaptive immune responses. It guides the development of lymphoid tissues and immune cells [58], affecting the movement of neutrophils and the division of T cells, especially regulatory T (Treg) cells to prevent aberrant inflammatory responses [2]. In a healthy gut, innate immune cells such as macrophages and dendritic cells capture luminal antigens and maintain tolerance by activating Treg cells rather than initiating a proinflammatory state [66]. These innate immune cells also migrate to lymphoid tissues to guide the adaptive response [66]. Immune defects have been found in germ-free (GF) mice where there is an absence of bacterial exposure. These defects include impaired development of gut-associated lymphoid tissues and Treg cells and reduced IgA-producing B cells [67,68].
The metabolic actions of the microbiome also contribute to immunity. Bacteria can assist in deriving energy from the fermentation of indigestible carbohydrates into short chain fatty acids (SCFAs), including acetate, propionate, and butyrate [69]. SCFAs can prevent the accumulation of toxic waste products [2,58] and provide chemo-protective properties for the intestinal barrier [3]. They maintain hypoxia, produce antimicrobial compounds, and promote anti-inflammatory cytokines. SCFAs also maintain the intestinal barrier [70] by promoting Treg cell development and enhancing mucous production from goblet cells [71].
However, a damaged barrier can allow bacteria and toxins to translocate into the systemic circulation [65], although the overall impact of translocated bacteria and toxins is dependent of the composition and types of bacteria in the intestinal luminal microbiome.

6. Characterising A Pathogenic Microbiome

6.1. Dysbiosis

Deviation from a healthy microbiome is commonly described as dysbiosis [66]. Dysbiosis can be defined as an altered composition and diversity of the gut microbiome that is associated with ill-health and disease. More specifically, dysbiosis disrupts immune suppressing host–microbe interactions and promotes inflammation, pathogenicity, and disease. With prolonged dysbiosis, there is an overgrowth of pathogenic bacteria with virulence factors, decreased regulatory or beneficial bacteria, and loss of bacterial diversity [72], which leads to dysfunction in the immune response and the promotion of a proinflammatory environment [65,66].
With dysbiosis, the homeostatic bacterial metabolic functions are altered, which can lead to a defective mucosal barrier [65]. It has been proposed that an increase in mucolytic bacteria such as Ruminococcus gnavus [73] and reduction in antimicrobial peptides such as Paneth cell-derived alpha-defensins, compromise the integrity of the mucosal barrier and are contributing factors in conditions such as inflammatory bowel disease (IBD) [74]. These factors also increase the risk of microbial overgrowth and bacterial translocation, which can further activate the host immune response via modulation of proinflammatory signalling pathways [65].
Pathobionts, or bacteria that display pathogenic properties opportunistically, can express virulence factors [75]. One example is the release of endotoxins, which act as ligands that bind cell surface receptors on innate immune cells, initiating cell activation [76]. Activated immune cells then upregulate their expression of pattern recognition receptors, which recognise microbe, pathogen, and danger-associated molecular patterns released by microbes and inflammatory cells [66]. In conjunction with reduced immune suppression from commensal bacteria, pathobionts lead to dysregulation in T cell differentiation that promotes the release of proinflammatory cytokines [77]. The resulting inflammatory environment induces host tissue damage and predisposes to diseases including IBD, irritable bowel syndrome, colorectal cancer, and extraintestinal diseases such as obesity and diabetes [78,79,80]. Table 1 outlines examples of several pathobionts and their molecular mediators suggested to contribute to disease.
A number of indices have been proposed to measure and assess dysbiosis. These include large-scale bacterial marker profiling, relevant taxa-based methods, neighborhood classification, random forest prediction, and combined alpha-beta diversity [89,90,91]. However, the most common method to measure and assess dysbiosis is to measure diversity. Alpha diversity describes the amount of unique taxa (richness) and their distribution (evenness) within a microbial community. Beta diversity is used to assess differences in microbial community composition between individuals, such as between those with a disease state and healthy controls [89]. Overall, multiple indices suggest that a single measure fails to address the broad concepts within dysbiosis. Furthermore, these measures cannot distinguish whether dysbiosis is a cause or consequence of disease [59,92,93].

6.2. The Pathogenic Microbiome in Inflammatory Disease

A prominent inflammatory disease is IBD, which is a chronic inflammatory condition of the GI tract with episodes of relapse and remission [65]; it is predominantly classified into Crohn’s disease (CD) and ulcerative colitis (UC). CD can affect the entire GI tract with transmural involvement, while UC is characterised by a more superficial inflammation that is confined to the large intestine [94]. The prevalence of IBD has increased in Western countries, with a rising incidence in newly industrialised countries [71]. Up to 25% of patients present with symptoms during adolescence and young adulthood [95]. Children typically develop a more aggressive disease course [96], with detrimental effects on growth, development, and psychosocial wellbeing [71]. The pathogenesis of IBD is multifactorial, with interactions between host genetics, a dysregulated immune response, environmental exposures, and importantly, changes in the gut microbiome contributing to disease [65].
Different factors can alter the intestinal microbiome [70]. Diet, including feeding from birth, is a notable environmental factor that profoundly impacts microbiome composition. For instance, the microbiome of formula-fed infants is characterised by decreased diversity and bacterial richness, which is associated with inflammation [97]. Bolte et al. [98] also found that the Westernised diet, consisting of lower dietary fibre and higher consumption of animal fat, salt, and sugar [99], induces microbiome characteristics that correspond with intestinal inflammation, while plant-based diets were associated with SCFA-producers and lower abundance of pathogenic bacteria. These observations are also supported by Zheng et al. [100] who found correlations between the abundance of several bacterial species and a proinflammatory diet (defined as a diet containing dietary components known to be associated with inflammation). Two further studies have also found associations between IBD and the consumption of high-sugar foods [101,102].
Antibiotic exposure is an additional environmental factor that can induce a loss of health-promoting bacteria and reduced expression of antibacterial agents and immunoglobulins, resulting in the potential to increase susceptibility to infections [55]. Antibiotic use corresponds with an increase in inflammatory cytokines, alteration of insulin sensitivity, and modulation of the metabolism of SCFAs and bile acids [103]. A cohort study conducted by Hviid et al. [104] reported associations between antibiotic exposure during childhood and the development of IBD. Additionally, maternal antibiotic use during pregnancy and infantile antibiotic exposure increases the risk of very early onset IBD [93,105]. Antibiotic use in infants small for gestational age showed an increase in pathogenic species associated with a ‘dysbiotic’ microbiome [13,106]. In addition to antibiotic exposure in premature and small-for-gestational-age infants, other early life factors including Caesarean section delivery and formula feeding increase the risks of asthma, atopy, obesity, type 2 diabetes mellitus, necrotising enterocolitis, and sepsis [5,6,13,36,37,107,108,109].
Significant efforts have been directed at identifying the pathogenic microbiome characteristics of inflammatory disease and in particular IBD [110,111,112,113,114,115,116]. Most studies have identified a decrease in alpha diversity in individuals with CD compared to UC and healthy controls [111,113,114,115,116]. However, a systematic review by Pittayanon et al. [110] reported that the most common findings of a decrease in the species Faecalibacterium prausnitzii and an increase in Enterobacteriaceae, were characteristic of IBD. F. prausnitzii is a SCFA-producer with metabolomic studies showing reduced SCFAs in the GI tract of individuals with IBD [112].
Furthermore, SCFA-producers, such as F. prausnitzii and Roseburia hominis, reduce proinflammatory cytokines including interleukin (IL)-12 and interferon-gamma and increase anti-inflammatory cytokines including IL-10 [59], implying that reduced SCFA-producing bacteria may be a marker of the pathogenic microbiome. Enterobacteriaceae, such as E. coli and Shigella, are facultative anaerobes and are enriched in inflammation at the expense of obligate anaerobes, which may be due to oxidative stress during inflammation [90]. These bacteria produce lipopolysaccharides (LPS), a pathogen-associated molecular pattern that activates Toll-like receptor signalling and a consequent inflammatory cascade [81].
As an illustration of this, a study of GF mice found that the mice developed colitis when colonised by a highly endotoxic intestinal microbiome containing a high proportion of Enterobacteriaceae (including E. coli) and a low proportion of Bacteroidetes [117]. Pathogenic strains of E. coli, including adherent-invasive E. coli (AIEC), have also been reported to be associated with IBD [118]. AIEC can induce the expression of cell adhesion molecules and they possess virulence factors such as type 1 pili and long polar fimbriae, which can facilitate colonisation in the intestinal mucosa [82]. Although E. coli can be considered part of the normal GI microbiome, a pathogenic and disproportionate abundance of E. coli may also constitute a pathogenic microbiome.
A caveat is that most studies on the microbiome in IBD are cross-sectional or longitudinal over short periods such as one year. Therefore, it is difficult to determine whether the previously described microbiome characteristics are a cause or consequence of disease. Furthermore, the GI microbiome is not homogenous, with reported differences in the microbiome between faecal and biopsy samples [81]. In addition, IBD is a heterogenous disease and findings for individuals with mild disease may not be valid for individuals with moderate or severe disease [111].

6.3. The Pediatric Pathogenic Microbiome

A further consideration of the contribution of the microbiome to disease is age. As previously described, in early childhood the gut microbiome is generally considered to be similar in composition and diversity to that of adults [119]. However, some studies report differences in the microbial composition between younger children and adolescents, suggesting continual development of the microbiome with age [62,97,120]. Furthermore, studies of the microbiome of treatment-naïve children with IBD suggests that the microbiome is characterised by reduced abundances of certain bacterial species rather than an increase in pathogens [63,90,121]. This highlights that in children at least, the pathogenic microbiome may be characterised by a loss of physiological functions.
However, a systematic review of gut microbiome profiles of children with IBD, commonly reported decreased alpha diversity and beta diversity that differed from healthy controls [71]. Overall, the IBD microbiome in children showed increased Enterococcus and decreased Anaerostipes, Blautia, Coprococcus, Faecalibacterium, Roseburia, Ruminococcus, and Lachnospira [71]. As these findings are also similar to adult IBD, it was proposed that the conditions for adult-onset IBD may be established during childhood [71].
Associations between the microbiome and disease severity or outcomes have also been investigated. Olbjørn et al. [120] reported that patients with higher abundance of Proteobacteria were more likely to require aggressive treatment and surgery. Proteobacteria abundance also correlated with increased CD complications and an absence of mucosal healing [120]. It is of interest that E. coli is classified within the Proteobacteria phylum. However, no firm conclusions can be reached whether specific species within the Proteobacteria phylum, or whether a common characteristic shared by the phylum, is contributing to these findings [120].
Paediatric studies have also investigated associations between dysbiosis, disease activity and treatment response [63,71,91,120,121,122,123]. Overall, dysbiosis positively correlated with disease activity in children [71,123]. Of interest was that hydrogen sulfide (H2S)-producers, such as Fusobacterium, Prevotella, and Streptococcus, were increased with inflammation [71]. H2S can damage the intestinal epithelium and influence the microbiome, mucous, and biofilm interactions [71]. Therefore, H2S and H2S-producers should also be a consideration of the pathogenic microbiome.
Contrary to these findings is that de Meij et al. [121] found no association between microbiome profile and disease activity. Further studies have also reported that with treatment, bacterial profiles and dysbiosis remain [91,120]. However, these studies also highlighted that consideration should also be given to the type of treatment, and/or treatment success. Kolho et al. [122] reported that microbial composition and diversity increased in treatment responders, following six weeks of anti-tumour necrosis factor-α therapy. Specific changes associated with treatment response have also been identified with Eubacterium and Bifidobacterium associated with a favorable response to medication [122,124]. A further study of the oral microbiome also found associations between bacterial species and treatment response [125]. These findings support the idea that analysis of the microbiome may potentially assist in the assessment and management of IBD.
A further factor that requires consideration is that the microbiome itself may be contributing to dysbiosis. The essence of this hypothesis is that symbiont bacteria can potentially evolve into pathobiont bacteria under specific selective pressures [126], which is feasible due to the rapid reproductive cycle of prokaryotes. The potential for a rapidly evolving microbiome is illustrated by the example that there are approximately the same number of reproductive events occurring in the average human microbiome in 5 days, as in the entire human population over the last 66 million years [127].
In recognition of these factors, the concept of microbiome engineering has emerged recently [128]. These include primary preventive strategies such as education on the consequences of Caesarean section delivery, conservative use of antibiotics during pregnancy, administration of antibiotics after cord clamping to limit foetal exposure and the adoption of the WHO Baby Friendly Hospital Initiative [9]. Current secondary prevention methods include pre- and probiotics supplementation of the mother during pregnancy and the neonate after birth, faecal microbiome transplantation and phage therapy [126]. At present, the literature surrounding neonatal microbiome engineering is relatively unexplored but poses a pivotal opportunity to promote robust microbiome development and prevent long-term pathologies associated with a disrupted microbiome.

7. Conclusions

Understanding what constitutes a heathy and diseased microbiome is in its infancy. Dysbiosis is increasingly used to describe a disease-causing or ‘pathogenic’ microbiome. However, dysbiosis remains an imprecise concept due to the substantial variability between healthy and diseased individuals, as well as variability in the diseases associated with dysbiosis. Nevertheless, there is some clarity regarding dysbiosis as a general concept. Dysbiosis can be regarded as a microbiome with diminished or lack of normal physiological functions, or one that favors inflammation over immune suppression. In this sense, dysbiosis can contribute to disease manifestation, rendering it pathogenic. Additionally, influential factors that contribute to a pathogenic microbiome are potentially in play even before birth. The importance of mother-to-infant vertical transfer of bacteria to the health of the infant is well-defined and underscores the significance of early microbiome development. Perturbations such as exposure to antibiotics and dietary choices, from birth and throughout life, have clear and measurable effects in altering the microbiome and contributing to dysbiosis.
Overall, the microbiome and its interaction with the host is a complex and highly variable ecosystem. However, investigating the microbiome and its dysbiosis potential (that is the functional, inflammatory, and anti-inflammatory capacity of the microbiome) opens the possibilities for predicting, diagnosing, managing, and potentially preventing inflammatory disease. This suggests a critical avenue for further research and novel interventions in the manipulation of the microbiome for improved human health.

Author Contributions

Conceptualization, S.T.L. and A.S.D. writing—original draft preparation, C.Y. and P.Y.W.; writing—review and editing, S.T.L., A.S.D. and D.A.L.; supervision, S.T.L. and D.A.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed]
  2. Afzaal, M.; Saeed, F.; Shah, Y.A.; Hussain, M.; Rabail, R.; Socol, C.T.; Hassoun, A.; Pateiro, M.; Lorenzo, J.M.; Rusu, A.V.; et al. Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 2022, 13, 999001. [Google Scholar] [CrossRef]
  3. Gomaa, E.Z. Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek 2020, 113, 2019–2040. [Google Scholar] [CrossRef]
  4. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed]
  5. Horta, B.L.; Loret de Mola, C.; Victora, C.G. Breastfeeding and intelligence: A systematic review and meta-analysis. Acta Paediatr. 2015, 104, 14–19. [Google Scholar] [CrossRef] [PubMed]
  6. Keag, O.E.; Norman, J.E.; Stock, S.J. Long-term risks and benefits associated with cesarean delivery for mother, baby, and subsequent pregnancies: Systematic review and meta-analysis. PLoS Med. 2018, 15, e1002494. [Google Scholar] [CrossRef]
  7. Wang, S.; Ryan, C.A.; Boyaval, P.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Maternal Vertical Transmission Affecting Early-life Microbiota Development. Trends Microbiol. 2020, 28, 28–45. [Google Scholar] [CrossRef]
  8. Ferretti, P.; Pasolli, E.; Tett, A.; Asnicar, F.; Gorfer, V.; Fedi, S.; Armanini, F.; Truong, D.T.; Manara, S.; Zolfo, M.; et al. Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 2018, 24, 133–145.e5. [Google Scholar] [CrossRef]
  9. Mueller, N.T.; Bakacs, E.; Combellick, J.; Grigoryan, Z.; Dominguez-Bello, M.G. The infant microbiome development: Mom matters. Trends Mol. Med. 2015, 21, 109–117. [Google Scholar] [CrossRef]
  10. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef]
  11. Gibson, M.K.; Wang, B.; Ahmadi, S.; Burnham, C.A.; Tarr, P.I.; Warner, B.B.; Dantas, G. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat. Microbiol. 2016, 1, 16024. [Google Scholar] [CrossRef] [PubMed]
  12. Harmsen, H.J.; Wildeboer-Veloo, A.C.; Raangs, G.C.; Wagendorp, A.A.; Klijn, N.; Bindels, J.G.; Welling, G.W. Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 61–67. [Google Scholar] [CrossRef] [PubMed]
  13. Mazzola, G.; Murphy, K.; Ross, R.P.; Di Gioia, D.; Biavati, B.; Corvaglia, L.T.; Faldella, G.; Stanton, C. Early Gut Microbiota Perturbations Following Intrapartum Antibiotic Prophylaxis to Prevent Group B Streptococcal Disease. PLoS ONE 2016, 11, e0157527. [Google Scholar] [CrossRef] [PubMed]
  14. Hay, W.W., Jr. The placenta. Not just a conduit for maternal fuels. Diabetes 1991, 40 (Suppl. S2), 44–50. [Google Scholar] [CrossRef]
  15. Lauder, A.P.; Roche, A.M.; Sherrill-Mix, S.; Bailey, A.; Laughlin, A.L.; Bittinger, K.; Leite, R.; Elovitz, M.A.; Parry, S.; Bushman, F.D. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome 2016, 4, 29. [Google Scholar] [CrossRef]
  16. Lager, S.; de Goffau, M.C.; Sovio, U.; Peacock, S.J.; Parkhill, J.; Charnock-Jones, D.S.; Smith, G.C.S. Detecting eukaryotic microbiota with single-cell sensitivity in human tissue. Microbiome 2018, 6, 151. [Google Scholar] [CrossRef]
  17. Lim, E.S.; Rodriguez, C.; Holtz, L.R. Reply Re: Amniotic fluid from healthy term pregnancies does not harbor a detectable microbial community. Microbiome 2019, 7, 21. [Google Scholar] [CrossRef]
  18. Theis, K.R.; Romero, R.; Winters, A.D.; Greenberg, J.M.; Gomez-Lopez, N.; Alhousseini, A.; Bieda, J.; Maymon, E.; Pacora, P.; Fettweis, J.M.; et al. Does the human placenta delivered at term have a microbiota? Results of cultivation, quantitative real-time PCR, 16S rRNA gene sequencing, and metagenomics. Am. J. Obstet. Gynecol. 2019, 220, 267.e1–267.e39. [Google Scholar] [CrossRef]
  19. Walker, R.W.; Clemente, J.C.; Peter, I.; Loos, R.J.F. The prenatal gut microbiome: Are we colonized with bacteria in utero? Pediatr. Obes. 2017, 12 (Suppl. S1), 3–17. [Google Scholar] [CrossRef]
  20. Rackaityte, E.; Halkias, J.; Fukui, E.M.; Mendoza, V.F.; Hayzelden, C.; Crawford, E.D.; Fujimura, K.E.; Burt, T.D.; Lynch, S.V. Viable bacterial colonization is highly limited in the human intestine in utero. Nat. Med. 2020, 26, 599–607. [Google Scholar] [CrossRef]
  21. Mishra, A.; Lai, G.C.; Yao, L.J.; Aung, T.T.; Shental, N.; Rotter-Maskowitz, A.; Shepherdson, E.; Singh, G.S.N.; Pai, R.; Shanti, A.; et al. Microbial exposure during early human development primes fetal immune cells. Cell 2021, 184, 3394–3409.e20. [Google Scholar] [CrossRef]
  22. Kelly, D.; King, T.; Aminov, R. Importance of microbial colonization of the gut in early life to the development of immunity. Mutat. Res. 2007, 622, 58–69. [Google Scholar] [CrossRef] [PubMed]
  23. Chu, D.M.; Ma, J.; Prince, A.L.; Antony, K.M.; Seferovic, M.D.; Aagaard, K.M. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 2017, 23, 314–326. [Google Scholar] [CrossRef] [PubMed]
  24. Rutayisire, E.; Huang, K.; Liu, Y.; Tao, F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: A systematic review. BMC Gastroenterol. 2016, 16, 86. [Google Scholar] [CrossRef] [PubMed]
  25. Shao, Y.; Forster, S.C.; Tsaliki, E.; Vervier, K.; Strang, A.; Simpson, N.; Kumar, N.; Stares, M.D.; Rodger, A.; Brocklehurst, P.; et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature 2019, 574, 117–121. [Google Scholar] [CrossRef]
  26. Chua, M.C.; Ben-Amor, K.; Lay, C.; Neo, A.G.E.; Chiang, W.C.; Rao, R.; Chew, C.; Chaithongwongwatthana, S.; Khemapech, N.; Knol, J.; et al. Effect of Synbiotic on the Gut Microbiota of Cesarean Delivered Infants: A Randomized, Double-blind, Multicenter Study. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 102–106. [Google Scholar] [CrossRef]
  27. Ronan, V.; Yeasin, R.; Claud, E.C. Childhood Development and the Microbiome-The Intestinal Microbiota in Maintenance of Health and Development of Disease during Childhood Development. Gastroenterology 2021, 160, 495–506. [Google Scholar] [CrossRef]
  28. Pannaraj, P.S.; Li, F.; Cerini, C.; Bender, J.M.; Yang, S.; Rollie, A.; Adisetiyo, H.; Zabih, S.; Lincez, P.J.; Bittinger, K.; et al. Association Between Breast Milk Bacterial Communities and Establishment and Development of the Infant Gut Microbiome. JAMA Pediatr. 2017, 171, 647–654. [Google Scholar] [CrossRef]
  29. Jost, T.; Lacroix, C.; Braegger, C.P.; Rochat, F.; Chassard, C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ. Microbiol. 2014, 16, 2891–2904. [Google Scholar] [CrossRef]
  30. Perez, P.F.; Doré, J.; Leclerc, M.; Levenez, F.; Benyacoub, J.; Serrant, P.; Segura-Roggero, I.; Schiffrin, E.J.; Donnet-Hughes, A. Bacterial imprinting of the neonatal immune system: Lessons from maternal cells? Pediatrics 2007, 119, e724–e732. [Google Scholar] [CrossRef]
  31. Cabrera-Rubio, R.; Collado, M.C.; Laitinen, K.; Salminen, S.; Isolauri, E.; Mira, A. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am. J. Clin. Nutr. 2012, 96, 544–551. [Google Scholar] [CrossRef]
  32. World Health Organisation. Infant and Young Child Feeding; World Health Organisation: Geneva, Switzerland, 2021.
  33. Rios-Leyvraz, M.; Yao, Q. The Volume of Breast Milk Intake in Infants and Young Children: A Systematic Review and Meta-Analysis. Breastfeed. Med. 2023, 18, 188–197. [Google Scholar] [CrossRef]
  34. Le Doare, K.; Holder, B.; Bassett, A.; Pannaraj, P.S. Mother’s Milk: A Purposeful Contribution to the Development of the Infant Microbiota and Immunity. Front. Immunol. 2018, 9, 361. [Google Scholar] [CrossRef]
  35. Carr, L.E.; Virmani, M.D.; Rosa, F.; Munblit, D.; Matazel, K.S.; Elolimy, A.A.; Yeruva, L. Role of Human Milk Bioactives on Infants’ Gut and Immune Health. Front. Immunol. 2021, 12, 604080. [Google Scholar] [CrossRef] [PubMed]
  36. Dogaru, C.M.; Nyffenegger, D.; Pescatore, A.M.; Spycher, B.D.; Kuehni, C.E. Breastfeeding and childhood asthma: Systematic review and meta-analysis. Am. J. Epidemiol. 2014, 179, 1153–1167. [Google Scholar] [CrossRef] [PubMed]
  37. Xu, L.; Lochhead, P.; Ko, Y.; Claggett, B.; Leong, R.W.; Ananthakrishnan, A.N. Systematic review with meta-analysis: Breastfeeding and the risk of Crohn’s disease and ulcerative colitis. Aliment. Pharmacol. Ther. 2017, 46, 780–789. [Google Scholar] [CrossRef]
  38. Asnicar, F.; Manara, S.; Zolfo, M.; Truong, D.T.; Scholz, M.; Armanini, F.; Ferretti, P.; Gorfer, V.; Pedrotti, A.; Tett, A.; et al. Studying Vertical Microbiome Transmission from Mothers to Infants by Strain-Level Metagenomic Profiling. mSystems 2017, 2, e00164-16. [Google Scholar] [CrossRef]
  39. Duranti, S.; Lugli, G.A.; Mancabelli, L.; Armanini, F.; Turroni, F.; James, K.; Ferretti, P.; Gorfer, V.; Ferrario, C.; Milani, C.; et al. Maternal inheritance of bifidobacterial communities and bifidophages in infants through vertical transmission. Microbiome 2017, 5, 66. [Google Scholar] [CrossRef]
  40. Kordy, K.; Gaufin, T.; Mwangi, M.; Li, F.; Cerini, C.; Lee, D.J.; Adisetiyo, H.; Woodward, C.; Pannaraj, P.S.; Tobin, N.H.; et al. Contributions to human breast milk microbiome and enteromammary transfer of Bifidobacterium breve. PLoS ONE 2020, 15, e0219633. [Google Scholar] [CrossRef] [PubMed]
  41. Milani, C.; Mancabelli, L.; Lugli, G.A.; Duranti, S.; Turroni, F.; Ferrario, C.; Mangifesta, M.; Viappiani, A.; Ferretti, P.; Gorfer, V.; et al. Exploring Vertical Transmission of Bifidobacteria from Mother to Child. Appl. Environ. Microbiol. 2015, 81, 7078–7087. [Google Scholar] [CrossRef]
  42. Biagi, E.; Aceti, A.; Quercia, S.; Beghetti, I.; Rampelli, S.; Turroni, S.; Soverini, M.; Zambrini, A.V.; Faldella, G.; Candela, M.; et al. Microbial Community Dynamics in Mother’s Milk and Infant’s Mouth and Gut in Moderately Preterm Infants. Front. Microbiol. 2018, 9, 2512. [Google Scholar] [CrossRef] [PubMed]
  43. Pärnänen, K.; Karkman, A.; Hultman, J.; Lyra, C.; Bengtsson-Palme, J.; Larsson, D.G.J.; Rautava, S.; Isolauri, E.; Salminen, S.; Kumar, H.; et al. Maternal gut and breast milk microbiota affect infant gut antibiotic resistome and mobile genetic elements. Nat. Commun. 2018, 9, 3891. [Google Scholar] [CrossRef] [PubMed]
  44. Wampach, L.; Heintz-Buschart, A.; Hogan, A.; Muller, E.E.L.; Narayanasamy, S.; Laczny, C.C.; Hugerth, L.W.; Bindl, L.; Bottu, J.; Andersson, A.F.; et al. Colonization and Succession within the Human Gut Microbiome by Archaea, Bacteria, and Microeukaryotes during the First Year of Life. Front. Microbiol. 2017, 8, 738. [Google Scholar] [CrossRef]
  45. Koenig, J.E.; Spor, A.; Scalfone, N.; Fricker, A.D.; Stombaugh, J.; Knight, R.; Angenent, L.T.; Ley, R.E. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4578–4585. [Google Scholar] [CrossRef] [PubMed]
  46. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef] [PubMed]
  47. Asakuma, S.; Hatakeyama, E.; Urashima, T.; Yoshida, E.; Katayama, T.; Yamamoto, K.; Kumagai, H.; Ashida, H.; Hirose, J.; Kitaoka, M. Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J. Biol. Chem. 2011, 286, 34583–34592. [Google Scholar] [CrossRef]
  48. Arrieta, M.C.; Stiemsma, L.T.; Amenyogbe, N.; Brown, E.M.; Finlay, B. The intestinal microbiome in early life: Health and disease. Front. Immunol. 2014, 5, 427. [Google Scholar] [CrossRef]
  49. Cheng, J.; Ringel-Kulka, T.; Heikamp-de Jong, I.; Ringel, Y.; Carroll, I.; de Vos, W.M.; Salojärvi, J.; Satokari, R. Discordant temporal development of bacterial phyla and the emergence of core in the fecal microbiota of young children. ISME J. 2016, 10, 1002–1014. [Google Scholar] [CrossRef]
  50. Kuang, Y.S.; Li, S.H.; Guo, Y.; Lu, J.H.; He, J.R.; Luo, B.J.; Jiang, F.J.; Shen, H.; Papasian, C.J.; Pang, H.; et al. Composition of gut microbiota in infants in China and global comparison. Sci. Rep. 2016, 6, 36666. [Google Scholar] [CrossRef]
  51. Adlerberth, I.; Jalil, F.; Carlsson, B.; Mellander, L.; Hanson, L.A.; Larsson, P.; Khalil, K.; Wold, A.E. High turnover rate of Escherichia coli strains in the intestinal flora of infants in Pakistan. Epidemiol. Infect. 1998, 121, 587–598. [Google Scholar] [CrossRef]
  52. Adlerberth, I.; Lindberg, E.; Aberg, N.; Hesselmar, B.; Saalman, R.; Strannegård, I.L.; Wold, A.E. Reduced enterobacterial and increased staphylococcal colonization of the infantile bowel: An effect of hygienic lifestyle? Pediatr. Res. 2006, 59, 96–101. [Google Scholar] [CrossRef] [PubMed]
  53. Huang, B.; Fettweis, J.M.; Brooks, J.P.; Jefferson, K.K.; Buck, G.A. The changing landscape of the vaginal microbiome. Clin. Lab. Med. 2014, 34, 747–761. [Google Scholar] [CrossRef] [PubMed]
  54. Neves, P.A.R.; Vaz, J.S.; Maia, F.S.; Baker, P.; Gatica-Domínguez, G.; Piwoz, E.; Rollins, N.; Victora, C.G. Rates and time trends in the consumption of breastmilk, formula, and animal milk by children younger than 2 years from 2000 to 2019: Analysis of 113 countries. Lancet Child Adolesc. Health 2021, 5, 619–630. [Google Scholar] [CrossRef] [PubMed]
  55. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef]
  56. Belizario, J.E.; Faintuch, J. Microbiome and Gut Dysbiosis. Exp. Suppl. 2018, 109, 459–476. [Google Scholar] [CrossRef]
  57. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef]
  58. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  59. Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome—A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [Google Scholar] [CrossRef]
  60. Yan, J.; Herzog, J.W.; Tsang, K.; Brennan, C.A.; Bower, M.A.; Garrett, W.S.; Sartor, B.R.; Aliprantis, A.O.; Charles, J.F. Gut microbiota induce IGF-1 and promote bone formation and growth. Proc. Natl. Acad. Sci. USA 2016, 113, E7554–E7563. [Google Scholar] [CrossRef]
  61. Ruan, W.; Engevik, M.A.; Spinler, J.K.; Versalovic, J. Healthy Human Gastrointestinal Microbiome: Composition and Function After a Decade of Exploration. Dig. Dis. Sci. 2020, 65, 695–705. [Google Scholar] [CrossRef]
  62. Hollister, E.B.; Riehle, K.; Luna, R.A.; Weidler, E.M.; Rubio-Gonzales, M.; Mistretta, T.A.; Raza, S.; Doddapaneni, H.V.; Metcalf, G.A.; Muzny, D.M.; et al. Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome 2015, 3, 36. [Google Scholar] [CrossRef]
  63. Kowalska-Duplaga, K.; Gosiewski, T.; Kapusta, P.; Sroka-Oleksiak, A.; Wedrychowicz, A.; Pieczarkowski, S.; Ludwig-Slomczynska, A.H.; Wolkow, P.P.; Fyderek, K. Differences in the intestinal microbiome of healthy children and patients with newly diagnosed Crohn’s disease. Sci. Rep. 2019, 9, 18880. [Google Scholar] [CrossRef] [PubMed]
  64. Alshehri, D.; Saadah, O.; Mosli, M.; Edris, S.; Alhindi, R.; Bahieldin, A. Dysbiosis of gut microbiota in inflammatory bowel disease: Current therapies and potential for microbiota-modulating therapeutic approaches. Bosn. J. Basic. Med. Sci. 2021, 21, 270–283. [Google Scholar] [CrossRef]
  65. Talapko, J.; Vcev, A.; Mestrovic, T.; Pustijanac, E.; Jukic, M.; Skrlec, I. Homeostasis and Dysbiosis of the Intestinal Microbiota: Comparing Hallmarks of a Healthy State with Changes in Inflammatory Bowel Disease. Microorganisms 2022, 10, 2405. [Google Scholar] [CrossRef] [PubMed]
  66. Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 3464. [Google Scholar] [CrossRef] [PubMed]
  67. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Nunez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
  68. Wang, H.; Wei, C.-X.; Min, L.; Zhu, L.-Y. Good or bad: Gut bacteria in human health and diseases. Biotechnol. Biotechnol. Equip. 2018, 32, 1075–1080. [Google Scholar] [CrossRef]
  69. Durda-Masny, M.; Ciomborowska-Basheer, J.; Makalowska, I.; Szwed, A. The Mediating Role of the Gut Microbiota in the Physical Growth of Children. Life 2022, 12, 152. [Google Scholar] [CrossRef]
  70. Kansal, S.; Catto-Smith, A.G.; Boniface, K.; Thomas, S.; Cameron, D.J.; Oliver, M.; Alex, G.; Kirkwood, C.D.; Wagner, J. The Microbiome in Paediatric Crohn’s Disease-A Longitudinal, Prospective, Single-Centre Study. J. Crohn’s Colitis 2019, 13, 1044–1054. [Google Scholar] [CrossRef]
  71. Zhuang, X.; Liu, C.; Zhan, S.; Tian, Z.; Li, N.; Mao, R.; Zeng, Z.; Chen, M. Gut Microbiota Profile in Pediatric Patients with Inflammatory Bowel Disease: A Systematic Review. Front. Pediatr. 2021, 9, 626232. [Google Scholar] [CrossRef]
  72. Eun, C.S.; Mishima, Y.; Wohlgemuth, S.; Liu, B.; Bower, M.; Carroll, I.M.; Sartor, R.B. Induction of bacterial antigen-specific colitis by a simplified human microbiota consortium in gnotobiotic interleukin-10-/- mice. Infect. Immun. 2014, 82, 2239–2246. [Google Scholar] [CrossRef]
  73. Png, C.W.; Lindén, S.K.; Gilshenan, K.S.; Zoetendal, E.G.; McSweeney, C.S.; Sly, L.I.; McGuckin, M.A.; Florin, T.H. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am. J. Gastroenterol. 2010, 105, 2420–2428. [Google Scholar] [CrossRef] [PubMed]
  74. Wehkamp, J.; Salzman, N.H.; Porter, E.; Nuding, S.; Weichenthal, M.; Petras, R.E.; Shen, B.; Schaeffeler, E.; Schwab, M.; Linzmeier, R.; et al. Reduced Paneth cell α-defensins in ileal Crohn’s disease. Proc. Natl. Acad. Sci. USA 2005, 102, 18129–18134. [Google Scholar] [CrossRef] [PubMed]
  75. Manos, J. The human microbiome in disease and pathology. APMIS 2022, 130, 690–705. [Google Scholar] [CrossRef]
  76. Lal, S.; Kandiyal, B.; Ahuja, V.; Takeda, K.; Das, B. Gut microbiome dysbiosis in inflammatory bowel disease. Prog. Mol. Biol. Transl. Sci. 2022, 192, 179–204. [Google Scholar] [CrossRef]
  77. Lee, M.; Chang, E.B. Inflammatory Bowel Diseases (IBD) and the Microbiome-Searching the Crime Scene for Clues. Gastroenterology 2021, 160, 524–537. [Google Scholar] [CrossRef]
  78. Ağagündüz, D.; Cocozza, E.; Cemali, Ö.; Bayazıt, A.D.; Nanì, M.F.; Cerqua, I.; Morgillo, F.; Saygılı, S.K.; Berni Canani, R.; Amero, P.; et al. Understanding the role of the gut microbiome in gastrointestinal cancer: A review. Front. Pharmacol. 2023, 14, 1130562. [Google Scholar] [CrossRef]
  79. Simon, D.; Kellermayer, R. Disturbed Pediatric Gut Microbiome Maturation in the Developmental Origins of Subsequent Chronic Disease. J. Pediatr. Gastroenterol. Nutr. 2023, 76, 123–127. [Google Scholar] [CrossRef]
  80. de Vos, W.M.; Tilg, H.; Van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 71, 1020–1032. [Google Scholar] [CrossRef] [PubMed]
  81. Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef]
  82. Azimi, T.; Nasiri, M.J.; Chirani, A.S.; Pouriran, R.; Dabiri, H. The role of bacteria in the inflammatory bowel disease development: A narrative review. APMIS 2018, 126, 275–283. [Google Scholar] [CrossRef]
  83. Smits, W.K.; Lyras, D.; Lacy, D.B.; Wilcox, M.H.; Kuijper, E.J. Clostridium difficile infection. Nat. Rev. Dis. Primers 2016, 2, 16020. [Google Scholar] [CrossRef]
  84. Wu, S.; Rhee, K.J.; Albesiano, E.; Rabizadeh, S.; Wu, X.; Yen, H.R.; Huso, D.L.; Brancati, F.L.; Wick, E.; McAllister, F.; et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 2009, 15, 1016–1022. [Google Scholar] [CrossRef]
  85. Goodwin, A.C.; Destefano Shields, C.E.; Wu, S.; Huso, D.L.; Wu, X.; Murray-Stewart, T.R.; Hacker-Prietz, A.; Rabizadeh, S.; Woster, P.M.; Sears, C.L.; et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 15354–15359. [Google Scholar] [CrossRef] [PubMed]
  86. Aviles-Jimenez, F.; Vazquez-Jimenez, F.; Medrano-Guzman, R.; Mantilla, A.; Torres, J. Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci. Rep. 2014, 4, 4202. [Google Scholar] [CrossRef] [PubMed]
  87. Seishima, J.; Iida, N.; Kitamura, K.; Yutani, M.; Wang, Z.; Seki, A.; Yamashita, T.; Sakai, Y.; Honda, M.; Yamashita, T.; et al. Gut-derived Enterococcus faecium from ulcerative colitis patients promotes colitis in a genetically susceptible mouse host. Genome Biol. 2019, 20, 252. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, J.; Hoedt, E.C.; Liu, Q.; Berendsen, E.; Teh, J.J.; Hamilton, A.; AW, O.B.; Ching, J.Y.L.; Wei, H.; Yang, K.; et al. Elucidation of Proteus mirabilis as a Key Bacterium in Crohn’s Disease Inflammation. Gastroenterology 2021, 160, 317–330.e11. [Google Scholar] [CrossRef] [PubMed]
  89. Wei, S.; Bahl, M.I.; Baunwall, S.M.D.; Hvas, C.L.; Licht, T.R. Determining Gut Microbial Dysbiosis: A Review of Applied Indexes for Assessment of Intestinal Microbiota Imbalances. Appl. Environ. Microbiol. 2021, 87, e00395-21. [Google Scholar] [CrossRef]
  90. Gevers, D.; Kugathasan, S.; Denson, L.A.; Vazquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef]
  91. Shaw, K.A.; Bertha, M.; Hofmekler, T.; Chopra, P.; Vatanen, T.; Srivatsa, A.; Prince, J.; Kumar, A.; Sauer, C.; Zwick, M.E.; et al. Dysbiosis, inflammation, and response to treatment: A longitudinal study of pediatric subjects with newly diagnosed inflammatory bowel disease. Genome Med. 2016, 8, 75. [Google Scholar] [CrossRef]
  92. Nagao-Kitamoto, H.; Shreiner, A.B.; Gillilland, M.G., 3rd; Kitamoto, S.; Ishii, C.; Hirayama, A.; Kuffa, P.; El-Zaatari, M.; Grasberger, H.; Seekatz, A.M.; et al. Functional Characterization of Inflammatory Bowel Disease-Associated Gut Dysbiosis in Gnotobiotic Mice. Cell. Mol. Gastroenterol. Hepatol. 2016, 2, 468–481. [Google Scholar] [CrossRef]
  93. Mark-Christensen, A.; Lange, A.; Erichsen, R.; Froslev, T.; Esen, B.O.; Sorensen, H.T.; Kappelman, M.D. Early-Life Exposure to Antibiotics and Risk for Crohn’s Disease: A Nationwide Danish Birth Cohort Study. Inflamm. Bowel Dis. 2022, 28, 415–422. [Google Scholar] [CrossRef] [PubMed]
  94. Fitzgerald, R.S.; Sanderson, I.R.; Claesson, M.J. Paediatric Inflammatory Bowel Disease and its Relationship with the Microbiome. Microb. Ecol. 2021, 82, 833–844. [Google Scholar] [CrossRef]
  95. Hoyhtya, M.; Korpela, K.; Saqib, S.; Junkkari, S.; Nissila, E.; Nikkonen, A.; Dikareva, E.; Salonen, A.; de Vos, W.M.; Kolho, K.L. Quantitative Fecal Microbiota Profiles Relate to Therapy Response during Induction with Tumor Necrosis Factor alpha Antagonist Infliximab in Pediatric Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2023, 29, 116–124. [Google Scholar] [CrossRef] [PubMed]
  96. Kugathasan, S.; Denson, L.A.; Walters, T.D.; Kim, M.O.; Marigorta, U.M.; Schirmer, M.; Mondal, K.; Liu, C.; Griffiths, A.; Noe, J.D.; et al. Prediction of complicated disease course for children newly diagnosed with Crohn’s disease: A multicentre inception cohort study. Lancet 2017, 389, 1710–1718. [Google Scholar] [CrossRef]
  97. Ihekweazu, F.D.; Versalovic, J. Development of the Pediatric Gut Microbiome: Impact on Health and Disease. Am. J. Med. Sci. 2018, 356, 413–423. [Google Scholar] [CrossRef] [PubMed]
  98. Bolte, L.A.; Vich Vila, A.; Imhann, F.; Collij, V.; Gacesa, R.; Peters, V.; Wijmenga, C.; Kurilshikov, A.; Campmans-Kuijpers, M.J.E.; Fu, J.; et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut 2021, 70, 1287–1298. [Google Scholar] [CrossRef] [PubMed]
  99. Yan, J.; Wang, L.; Gu, Y.; Hou, H.; Liu, T.; Ding, Y.; Cao, H. Dietary Patterns and Gut Microbiota Changes in Inflammatory Bowel Disease: Current Insights and Future Challenges. Nutrients 2022, 14, 4003. [Google Scholar] [CrossRef]
  100. Zheng, J.; Hoffman, K.L.; Chen, J.S.; Shivappa, N.; Sood, A.; Browman, G.J.; Dirba, D.D.; Hanash, S.; Wei, P.; Hebert, J.R.; et al. Dietary inflammatory potential in relation to the gut microbiome: Results from a cross-sectional study. Br. J. Nutr. 2020, 124, 931–942. [Google Scholar] [CrossRef]
  101. Peters, V.; Tigchelaar-Feenstra, E.F.; Imhann, F.; Dekens, J.A.M.; Swertz, M.A.; Franke, L.H.; Wijmenga, C.; Weersma, R.K.; Alizadeh, B.Z.; Dijkstra, G.; et al. Habitual dietary intake of IBD patients differs from population controls: A case-control study. Eur. J. Nutr. 2021, 60, 345–356. [Google Scholar] [CrossRef]
  102. Racine, A.; Carbonnel, F.; Chan, S.S.; Hart, A.R.; Bueno-de-Mesquita, H.B.; Oldenburg, B.; van Schaik, F.D.; Tjonneland, A.; Olsen, A.; Dahm, C.C.; et al. Dietary Patterns and Risk of Inflammatory Bowel Disease in Europe: Results from the EPIC Study. Inflamm. Bowel Dis. 2016, 22, 345–354. [Google Scholar] [CrossRef]
  103. Vandenplas, Y.; Carnielli, V.P.; Ksiazyk, J.; Luna, M.S.; Migacheva, N.; Mosselmans, J.M.; Picaud, J.C.; Possner, M.; Singhal, A.; Wabitsch, M. Factors affecting early-life intestinal microbiota development. Nutrition 2020, 78, 110812. [Google Scholar] [CrossRef] [PubMed]
  104. Hviid, A.; Svanstrom, H.; Frisch, M. Antibiotic use and inflammatory bowel diseases in childhood. Gut 2011, 60, 49–54. [Google Scholar] [CrossRef]
  105. Örtqvist, A.K.; Lundholm, C.; Halfvarson, J.; Ludvigsson, J.F.; Almqvist, C. Fetal and early life antibiotics exposure and very early onset inflammatory bowel disease: A population-based study. Gut 2019, 68, 218–225. [Google Scholar] [CrossRef]
  106. Nogacka, A.; Salazar, N.; Suárez, M.; Milani, C.; Arboleya, S.; Solís, G.; Fernández, N.; Alaez, L.; Hernández-Barranco, A.M.; de Los Reyes-Gavilán, C.G.; et al. Impact of intrapartum antimicrobial prophylaxis upon the intestinal microbiota and the prevalence of antibiotic resistance genes in vaginally delivered full-term neonates. Microbiome 2017, 5, 93. [Google Scholar] [CrossRef]
  107. Riva, A.; Borgo, F.; Lassandro, C.; Verduci, E.; Morace, G.; Borghi, E.; Berry, D. Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ. Microbiol. 2017, 19, 95–105. [Google Scholar] [CrossRef]
  108. Ahmadizar, F.; Vijverberg, S.J.H.; Arets, H.G.M.; de Boer, A.; Lang, J.E.; Garssen, J.; Kraneveld, A.; Maitland-van der Zee, A.H. Early-life antibiotic exposure increases the risk of developing allergic symptoms later in life: A meta-analysis. Allergy 2018, 73, 971–986. [Google Scholar] [CrossRef] [PubMed]
  109. Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Nikolopoulos, G.K.; Lytras, T.; Bonovas, S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Meta-analyses. Gastroenterology 2019, 157, 647–659.e4. [Google Scholar] [CrossRef] [PubMed]
  110. Pittayanon, R.; Lau, J.T.; Leontiadis, G.I.; Tse, F.; Yuan, Y.; Surette, M.; Moayyedi, P. Differences in Gut Microbiota in Patients With vs Without Inflammatory Bowel Diseases: A Systematic Review. Gastroenterology 2020, 158, 930–946.e1. [Google Scholar] [CrossRef] [PubMed]
  111. Amos, G.C.A.; Sergaki, C.; Logan, A.; Iriarte, R.; Bannaga, A.; Chandrapalan, S.; Wellington, E.M.H.; Rijpkema, S.; Arasaradnam, R.P. Exploring how microbiome signatures change across inflammatory bowel disease conditions and disease locations. Sci. Rep. 2021, 11, 18699. [Google Scholar] [CrossRef]
  112. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
  113. Gallagher, K.; Catesson, A.; Griffin, J.L.; Holmes, E.; Williams, H.R.T. Metabolomic Analysis in Inflammatory Bowel Disease: A Systematic Review. J. Crohn’s Colitis 2021, 15, 813–826. [Google Scholar] [CrossRef]
  114. Serrano-Gomez, G.; Mayorga, L.; Oyarzun, I.; Roca, J.; Borruel, N.; Casellas, F.; Varela, E.; Pozuelo, M.; Machiels, K.; Guarner, F.; et al. Dysbiosis and relapse-related microbiome in inflammatory bowel disease: A shotgun metagenomic approach. Comput. Struct. Biotechnol. J. 2021, 19, 6481–6489. [Google Scholar] [CrossRef]
  115. Pascal, V.; Pozuelo, M.; Borruel, N.; Casellas, F.; Campos, D.; Santiago, A.; Martinez, X.; Varela, E.; Sarrabayrouse, G.; Machiels, K.; et al. A microbial signature for Crohn’s disease. Gut 2017, 66, 813–822. [Google Scholar] [CrossRef] [PubMed]
  116. Alam, M.T.; Amos, G.C.A.; Murphy, A.R.J.; Murch, S.; Wellington, E.M.H.; Arasaradnam, R.P. Microbial imbalance in inflammatory bowel disease patients at different taxonomic levels. Gut Pathog. 2020, 12, 1. [Google Scholar] [CrossRef]
  117. Gronbach, K.; Flade, I.; Holst, O.; Lindner, B.; Ruscheweyh, H.J.; Wittmann, A.; Menz, S.; Schwiertz, A.; Adam, P.; Stecher, B.; et al. Endotoxicity of Lipopolysaccharide as a Determinant of T-Cell−Mediated Colitis Induction in Mice. Gastroenterology 2014, 146, 765–775. [Google Scholar] [CrossRef] [PubMed]
  118. Petersen, A.M.; Halkjær, S.I.; Gluud, L.L. Intestinal colonization with phylogenetic group B2 Escherichia coli related to inflammatory bowel disease: A systematic review and meta-analysis. Scand. J. Gastroenterol. 2015, 50, 1199–1207. [Google Scholar] [CrossRef]
  119. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  120. Olbjørn, C.; Cvancarova Småstuen, M.; Thiis-Evensen, E.; Nakstad, B.; Vatn, M.H.; Jahnsen, J.; Ricanek, P.; Vatn, S.; Moen, A.E.F.; Tannæs, T.M.; et al. Fecal microbiota profiles in treatment-naïve pediatric inflammatory bowel disease—Associations with disease phenotype, treatment, and outcome. Clin. Exp. Gastroenterol. 2019, 12, 37–49. [Google Scholar] [CrossRef]
  121. de Meij, T.G.J.; de Groot, E.F.J.; Peeters, C.F.W.; de Boer, N.K.H.; Kneepkens, C.M.F.; Eck, A.; Benninga, M.A.; Savelkoul, P.H.M.; van Bodegraven, A.A.; Budding, A.E. Variability of core microbiota in newly diagnosed treatment-naive paediatric inflammatory bowel disease patients. PLoS ONE 2018, 13, e0197649. [Google Scholar] [CrossRef]
  122. Kolho, K.L.; Korpela, K.; Jaakkola, T.; Pichai, M.V.; Zoetendal, E.G.; Salonen, A.; de Vos, W.M. Fecal Microbiota in Pediatric Inflammatory Bowel Disease and Its Relation to Inflammation. Am. J. Gastroenterol. 2015, 110, 921–930. [Google Scholar] [CrossRef]
  123. Malham, M.; Lilje, B.; Houen, G.; Winther, K.; Andersen, P.S.; Jakobsen, C. The microbiome reflects diagnosis and predicts disease severity in paediatric onset inflammatory bowel disease. Scand. J. Gastroenterol. 2019, 54, 969–975. [Google Scholar] [CrossRef]
  124. Wang, Y.; Gao, X.; Zhang, X.; Xiao, F.; Hu, H.; Li, X.; Dong, F.; Sun, M.; Xiao, Y.; Ge, T.; et al. Microbial and metabolic features associated with outcome of infliximab therapy in pediatric Crohn’s disease. Gut Microbes 2021, 13, 1865708. [Google Scholar] [CrossRef]
  125. Elmaghrawy, K.; Fleming, P.; Fitzgerald, K.; Cooper, S.; Dominik, A.; Hussey, S.; Moran, G.P. The Oral Microbiome in Treatment-Naïve Paediatric IBD Patients Exhibits Dysbiosis Related to Disease Severity that Resolves following Therapy. J. Crohn’s Colitis 2022, 17, 553–564. [Google Scholar] [CrossRef] [PubMed]
  126. Bliven, K.A.; Maurelli, A.T. Evolution of Bacterial Pathogens Within the Human Host. Microbiol. Spectr. 2016, 4, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  127. Venkatakrishnan, A.; Holzknecht, Z.E.; Holzknecht, R.; Bowles, D.E.; Kotzé, S.H.; Modliszewski, J.L.; Parker, W. Evolution of bacteria in the human gut in response to changing environments: An invisible player in the game of health. Comput. Struct. Biotechnol. J. 2021, 19, 752–758. [Google Scholar] [CrossRef] [PubMed]
  128. Wong, E.; Lui, K.; Day, A.S.; Leach, S.T. Manipulating the neonatal gut microbiome: Current understanding and future perspectives. Arch. Dis. Child. Fetal Neonatal Ed. 2022, 107, 346–350. [Google Scholar] [CrossRef] [PubMed]
Table 1. Examples of pathobionts and their molecular mediators associated with disease.
Table 1. Examples of pathobionts and their molecular mediators associated with disease.
PathobiontsMolecular MediatorsAssociation with Disease
Escherichia coliProduce lipopolysaccharides (LPS), activating toll-like receptor (TLR) signalling and inflammatory cascades including release of interleukin-8 (IL-8) in colonic and intestinal epithelial cells [81]
Adherent invasive E. coli strains express cell adhesion molecules to facilitate colonisation in the intestinal mucosa, with virulence factors including type 1 pili and long polar fimbriae expression [82]
Promote cancer through activation of T-helper (Th)-17 cells, direct DNA damage, and induction of cholesterol synthesis [78]
IBD
Type 2 Diabetes
Fatty liver disease
Colorectal cancer [3]
Clostridioides difficileMucolytic enzymes such as cell surface protein Cwp84 which degrades the colonic mucosa and surface layer protein A (SlpA) which activates TLR4-dependent responses and aids colonisation
Clostridial toxins A and B (TcdA and TcdB) stimulate inflammatory chemokine and cytokine production, neutrophil influx, disruption of tight junctions, fluid secretion, and epithelial cell death [83]
Pseudomembranous colitis
Enterotoxigenic Bacteroides fragilis (ETBF)Induces colonic signal transducer and activator of transcription-3 (Stat3) activation; colitis characterised by Th-17 response [84]
Induces spermine oxidase (SMO) resulting in increased reactive oxygen species (ROS) and DNA damage [85]
Colorectal cancer
Helicobacter pyloriInduces SMO leading to ROS production and DNA damage [85]
Cytotoxins VacA and CagA injected into host cell, activating oncogenic signal transduction pathways [78]
Changes in microbial flora from atrophic gastritis to intestinal metaplasia and dysplasia, leading to increased carcinogenic potential of bacterial strains and formation of carcinogenic N-nitroso compounds [86]
Peptic ulcer disease, gastritis, gastric cancer
Enterococcus faecalisInoculation of E. faecium strain ATCC 19434 in IL-10 depleted mice increased expression of inflammatory cytokines tumour necrosis factor, IL-1b, IL-6, IL-17a, and IL-12b [87]Ulcerative colitis
Proteus mirabilisP. mirabilis strains present in mice showed bacterial adherence, invasion, and increased production of pro-inflammatory cytokines IL-18 and IL-1α and cell necrosis
Induce pro-inflammatory pathways including nucleotide-binding oligomerisation domain (NOD)-like receptor signalling, Jak-STAT signalling, and MAPK signalling [88]
Crohn’s disease
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wong, P.Y.; Yip, C.; Lemberg, D.A.; Day, A.S.; Leach, S.T. Evolution of a Pathogenic Microbiome. J. Clin. Med. 2023, 12, 7184. https://doi.org/10.3390/jcm12227184

AMA Style

Wong PY, Yip C, Lemberg DA, Day AS, Leach ST. Evolution of a Pathogenic Microbiome. Journal of Clinical Medicine. 2023; 12(22):7184. https://doi.org/10.3390/jcm12227184

Chicago/Turabian Style

Wong, Pui Yin, Carmen Yip, Daniel A. Lemberg, Andrew S. Day, and Steven T. Leach. 2023. "Evolution of a Pathogenic Microbiome" Journal of Clinical Medicine 12, no. 22: 7184. https://doi.org/10.3390/jcm12227184

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop