Open access peer-reviewed chapter - ONLINE FIRST

The Intestinal Microbiome in Humans: Its Role for a Healthy Life and in the Onset of Diseases

Written By

Bogdan Severus Gaspar, Monica Profir, Oana Alexandra Rosu, Ruxandra Florentina Ionescu and Sanda Maria Cretoiu

Submitted: 22 August 2023 Reviewed: 12 October 2023 Published: 29 May 2024

DOI: 10.5772/intechopen.113719

Human Physiology - Annual Volume 2024 IntechOpen
Human Physiology - Annual Volume 2024 Authored by Kunihiro Sakuma

From the Annual Volume

Human Physiology - Annual Volume 2024 [Working Title]

Prof. Kunihiro Sakuma and Dr. Kotomi Sakai

Chapter metrics overview

37 Chapter Downloads

View Full Metrics

Abstract

The discovery of human microbiota shed a different perspective regarding human homeostasis and immune regulation. Gut microbiota comprises a multitude of microorganisms, its composition being host-specific and evolving throughout the lifetime, being subjected to both endogenous and exogenous factors. This subject gained significant interest after the improvement of metagenomic and metabolomic studies. The gut microbiome displays several roles, such as modulating gut permeability, digestive processes, metabolic pathways, and immune responses. Any dysregulation in the complex symbiosis mechanism between humans and the intestinal microbiome might lead to variable diseases. Environmental factors and diet play a very important role in maintaining a healthy gut microbiota. In this chapter, one aims to discuss the core microbiome of healthy subjects and how different stages of dysbiosis can play a role in the initiation and progression of pathogenic mechanisms leading to several diseases, such as gastrointestinal disorders (irritable bowel syndrome, inflammatory bowel diseases, infections or diarrhea associated with antibiotics, and colon cancer), metabolic disorders, obesity, diabetes, and allergies. We underline the importance of diet and environmental factors in modulating gut microorganism concentrations. We shed light on new possible perspectives regarding the modulation of gut microbiota for improving the health status of the host.

Keywords

  • intestinal microbiota
  • gut microbiome
  • dysbiosis
  • immune defense
  • healthy gut

1. Introduction

Although the terms “microbiota” and “microbiome” are frequently used interchangeably, there are some distinctions between the two. The term “microbiota” refers to the live microorganisms found in specific environments, such as the gut and oral microbiota. The term “microbiome” refers to collecting all genomes within the microbiota, along with microbial structural elements, metabolites, and environmental conditions [1].

More than 100 trillion microorganisms, such as bacteria, yeast, and viruses, compose the human gut microbiota [2]. The gastrointestinal tract (GI) is considered to be the largest interface (250–400 m2) between the host, environmental factors, and antigens in the human body [2].

The composition of gut microbiota expands along the gastrointestinal tract, reaching its highest concentration at the colon level. This can be explained by the rare resistance of bacteria to the acidic medium or enzymes found in the stomach and proximal duodenum [3]. The bacterial density in the stomach is about 10 bacteria per gram, 103 per gram in the duodenum, 104 per gram in the jejunum, 107 per gram in the ileum, and 1012 per gram in the colon [4].

About 1000 bacterial species can be found in the colon, comprising approximately three million genes [5].

The bacteria that colonize the adult human gut are grouped into 7 major phyla: Actinobacteria, Bacteroidetes, Firmicutes, Fusobacteria, Euryarchaeota, Proteobacteria, and Verrucomicrobia. The dominant bacterial phyla are Firmicutes and Bacteroidetes, accounting for 90% of the gut microbiota [2, 6]. Moreover, the gut microbiome encodes over 100 times as many genes as the human genome [6]. The gut microbiome is often referred to as a “superorganism” due to its microbial diversity and its symbiotic relationship with the human body [7, 8]. In the last years, stool analysis techniques have facilitated researchers to analyze the components of the GI microbiota, with 16S ribosomal RNA gene sequencing being the most useful technique for describing gut microbial strains [6].

Gut bacteria is a key part of the digestion process. Commensal bacteria can extract and synthesize essential amino acids, vitamins, and short-chain fatty acids (SCFAs), helping digest dietary components that the human body alone cannot process, such as polysaccharides [9]. Intestinal bacteria also play a key role in maintaining immune and metabolic homeostasis and protecting the host against pathogen colonization [10].

Microorganisms interconnect with one another, having symbiotic reactions, generating various effects on population composition and activity within the gut microbiome. The interactions can be between microorganisms from the same species as well as from different ones. The interplay can be categorized as mutualism, synergism, commensalism, or, with a negative effect, amensalism (parasitism, antagonism, and competitiveness). Nevertheless, secondary metabolites are of high importance in maintaining a well-functioning ecosystem [1].

Colonic bacteria can digest complex carbohydrates and create metabolites like short-chain fatty acids (SCFAs) by producing enzymes that are active on carbohydrates [2]. Propionate, butyrate, and acetate are the three main SCFAs that are typically found in the gastrointestinal system in a ratio of 1:1:3 [2]. These SCFAs are swiftly absorbed by the enteral epithelial cells and are involved in the regulation of cellular processes such as gene expression, chemotaxis, differentiation, proliferation, and cellular death [2]. The gut microbiota is also implicated in the synthesis of essential vitamins, which the host is unable to manufacture [2]. Through its structural components and metabolites, the gut microbiota stimulates the host to produce various antimicrobial compounds [2]. These include AMPs like cathelicidins, C-type lectins, and prodefensins that are produced by the host Paneth cells via a pattern recognition receptor-mediated mechanism [2]. The other method through which the gut microbiota inhibits pathogen overgrowth is by inducing mucosal secretory immunoglobulin A [2].

The human microbiome is seen an essential therapeutic objective in the area of personalized medicine by providing fascinating therapeutic keys for a multitude of conditions. Its composition can be influenced by diet modulation, by adding prebiotics, probiotics, or synbiotics or by even taking into consideration microbiome transplants, of which fecal transplant is drawing more attention [1].

Intestinal microorganisms can influence the immune or metabolic pathways of several diseases, making the host vulnerable or immune to certain pathogens [2].

The relevance of a healthy gut microbiome can be reflected by the gut–brain, gut–lung, or even gut–liver axis, making the study in this field an increasingly alluring research topic [1].

There is growing evidence in the literature regarding the bidirectional connection between the human gut bacteria and the brain. The microbiota has effects on the central nervous system through neuroendocrine- and immune-mediated mechanisms, while the brain can act on the gut microbiota through the autonomic nervous system. SCFAs can modulate the intestinal barrier function, the secretion of mucosal neurotransmitters, the sympathetic nervous system, or the concentrations of neurotransmitters, such as GABA, serotonin, histamine, melatonin, and acetylcholine. Additionally, gut microbes can alter afferent sensory nerves by inhibiting calcium-dependent potassium channels. They can also have an impact on the role of immunity [3]. By regulation of gastrointestinal motility and mucus production and change of the intestinal permeability and immunological functions, the brain affects the gut microbiota [3].

This review aims to summarize our current understanding of the development of gut microbiota and the changes that arise during different stages and conditions.

Advertisement

2. The core microbiome of a healthy individual

The core microbiome refers to a set of stable components across populations. The taxa that compose the core microbiota must be present in most subjects in appreciable amounts [11]. However, both researchers and international projects strived for decades to define this set of components [12, 13]. The core microbiome comprises a group of strains that are transferred from the mother before birth and continue to evolve over the first few months of life based on the infant’s nutrition and environmental factors [14]. The ability of the core microbiome to resist changes brought on by perturbations and to return to an equilibrium state following concerns is one of its key characteristics [15]. This chapter aims to define the core microbiome and how it fluctuates across different age groups.

2.1 Early childhood development of the microbiome

There is conflicting evidence regarding whether or not the microbiome is influenced before birth by the microbial colonization of the placenta [16]. The presence of bacteria in the placenta, the amniotic cavity, the umbilical cord, and meconium has led some scientists to support the theory that infant gut colonization begins in utero [17, 18, 19, 20, 21, 22, 23, 24].

However, other studies agree that gut colonization starts at birth, and this microbial challenge leads to the development of the immune system [25]. Current data has demonstrated a significant increase in facultative anaerobes and obligate anaerobes after birth [26].

Studies suggest significant differences in intestinal microbial composition in infants born vaginally and infants delivered via C-section (Figure 1) [27].

Figure 1.

Factors contributing to the development of the microbiota in early life. Created with Biorender.com.

Vaginal delivery is associated with early colonization with microbes normally found in the vaginal tract, such as Lactobacillus, Prevotella, and Sneathia spp. [28]. The intestinal microbiome of infants born via C-section is influenced by bacteria typically found on the mother’s skin, oral microbiome bacteria originating from the mother’s stool, and even bacteria found on hospital surfaces [27, 29]. This leads to a flora dominated by Staphylococcus, Corynebacterium, and Propionibacterium [29]. Moreover, in the first months of life of infants born vaginally, there tends to be an increase in the proportion of Bifidobacteria and Bacteroides. In contrast, this proportion remains low in infants delivered via C-section [29, 30]. This is of great importance considering that both Bacteroides and Bifidobacteria are considered health-promoting bacteria [31]. On the other hand, increased numbers of potentially pathogenic, proinflammatory bacteria such as Klebsiella and Enterococcus seem to be present in the intestinal flora of children born by C-section [30, 32, 33]. In addition, studies have correlated these differences in the gut microbial composition with a predisposition to infections, allergies, and inflammatory diseases [28, 30, 34, 35].

Another factor that influences the gut microbiome development is the infant diet. Specifically, the intestinal microbiota of infants that are breast-fed is dominated by Lactobacillus, Staphylococcus, and Bifidobacterium. In contrast, the gut microbiome of formula-fed infants mainly consists of bacteria such as Roseburia, Clostridium, and Anaerosipes [16, 27, 36]. Interestingly, after breastfeeding cessation, the gut microbial composition rapidly swifts toward a composition closer to the adult microbial flora [27]. In preterm infants, delayed colonization and low diversity are representative for the intestinal microbiome. Moreover, some studies have shown that in preterm infants, the levels of potential pathogenic bacteria and facultative anaerobic bacteria are increased. In contrast, the levels of mandatory anaerobic bacteria seem to be decreased [37, 38, 39]. These differences in terms of microbial composition have been associated with several disorders such as gastrointestinal infections and necrotizing enterocolitis [40, 41].

In the first year of life, the infant’s gut microbiome matures and becomes more complex. This time period seems to depend on several environmental factors like antibiotic treatment, living with household pets, and growing up with siblings [36]. Antibiotic exposure leads to changes in the intestinal microbial population, such as a less diversity of the microbiota. It also increases the risk of allergies, asthma, and metabolic disorders [36, 42]. Between the ages of 1–3 years old, microbial diversity increases, and at 3 years old, it becomes more stable and resembles the composition of the adult gut microbiota [43].

2.2 The adult healthy microbiome diversity

The gut microbial community composition in adults is highly variable in the same individual and among individuals [44]. Microbial richness and diversity are characteristic of a healthy gut microbiome. The composition of the gut microbiome depends on age, sex, dietary habits, antibiotic use, physical activity, and weight.

Three bacterial phyla dominate the healthy adult gut microbiota, Firmicutes (Lachnospiraceae and Ruminococcaceae), Bacteroidetes (Bacteroidaceae, Prevotellaceae, and Rikenellaceae), and Actinobacteria (Bifidobacteriaceae and Coriobacteriaceae). The Firmicutes/Bacteroidetes (F/B) ratio plays a major role in maintaining homeostasis, and variations in this ratio have been correlated with several pathologies. An increased number of Firmicutes and a reduced population of Bacteroidetes have been observed in obese patients. In contrast, a decrease in the F/B ratio has been associated with the development of inflammatory bowel disease [45, 46].

The MetaHIT consortium proposed classifying the gut flora by enterotypes, Enterotype 1, characterized by a high abundance of Bacteroides; Enterotype 2, characterized by a high abundance of Prevotella; and Enterotype 3, which has a high concentration of Ruminococcus [47]. An enterotype represents a functional association of bacteria of different species with specific ways of generating energy from fermentable dietary components found in the colon [48]. Bacteria from enterotype 1 generate energy by degrading carbohydrates and proteins, and bacteria clusters of enterotypes 2 and 3 degrade mucin glycoproteins of the gut mucosal layer [48].

One of the challenges in defining a healthy microbiome comes from the fact that most studies on microbiome composition have included participants from socioeconomically developed countries from Europe and North America. The microbiomes of people from nonindustrialized and industrialized regions differ in composition, with a nonindustrialized microbiome being adapted to fiber degradation. In contrast, an industrialized microbiota is adapted for mucin degradation due to antibiotic and different drug exposure [49]. For example, the microbiota of Hadza hunter-gatherers of Tanzania is characterized by a greater phylogenetic and carbohydrate-active enzyme diversity than that of Western populations [50]. In addition, a study that compared the gut microbiota of 106 Japanese subjects with the gut microbiota of subjects from 11 different countries showed significant differences between the two groups. Namely, the Japanese gut microbiota is characterized by a higher abundance of the phylum Actinobacteria due to its high quantity of Bifidobacterium species, a remarkable depletion of the archaeon Methanobrevibacter smithii, and an overall depletion of genes responsible for methanogenesis and increased levels of anaerobic acetogens such as Blautia species. The Japanese gut microbiota also has a high carbohydrate metabolism capacity, which translates into higher levels of SCFAs and hydrogen end products, positively affecting host health [51].

Decreased microbial diversity has been correlated with several chronic illnesses, such as diabetes and obesity [52].

Increased diversity has been associated with longevity in some studies on Asian populations [53, 54]. Moreover, when comparing the gut microbiome of long-living Chinese patients to the microbiomes of a group of long-living Italians, it was demonstrated that although the groups differed in composition, diversity was the common characteristic. They also discovered that the long-living Chinese have higher concentrations of a particular bacterial genus, Clostridium cluster XIVa, which is renowned for producing SCFAs [52].

2.3 The microbiome in elderly and age-related dysbiosis

The gut microbiota changes substantially in diversity and microbial composition with age, especially in people over 70 years old [55]. Most of the elderly experience comorbidities associated with gut microbiota; thus, it is difficult to define the healthy gut microbiome in this age group.

The reduced microbial diversity and the differences in the gut microbial composition of the elderly compared to healthy adults have been attributed to lifestyle changes, dietary schedule, reduced mobility, weakened immune strength, decreased intestinal function, altered gut morphology and physiology, infections, and increased hospitalizations and medication [11, 52, 55, 56, 57, 58]. Reduced gut microbial diversity of the core microbial groups has been associated with increased frailty and reduced cognitive performance [59].

The gut microbiota of the elderly is generally characterized by reduced amounts of commensals like Bacteroides, Bifidobacteria, and Lactobacilli and increased amounts of opportunistic bacteria such as Clostridium perfingens, Clostridium difficile, and enterobacteria [11, 60, 61, 62, 63]. At a phylum level, the microbiota of elderly subjects is dominated by Bacteroidetes, while the microbiota of healthy adults is dominated by the Firmicutes phylum [55]. In addition, studies have demonstrated that increased levels of Proteobacteria are common in the microbiota of elderly and long-living subjects [54].

In addition, the microbiota of the elderly is characterized by a reduced metabolic capacity with lower SCFAs levels [64]. The lower levels of SCFAs are closely related to the so-called “inflamaging” process that appears in the intestine of aged people [65]. Compared to younger subjects, the intestinal bacterial metabolism shifts from predominantly saccharolytic to a predominantly putrefactive metabolism in older subjects. This might account for the changes in bacterial metabolism and the production of bacterial metabolites [66].

Numerous researchers have looked at the microbiota of centenarians to characterize the healthy microbiome of the elderly [53, 54, 58, 67]. This research set out to identify the distinctive features of the centenarian microbiome that enable them to adapt to the accumulating oxidative alterations and chronic inflammatory processes that define the extreme aging phenotype. These features are represented by increased diversity of microbial populations, and enriched populations of beneficial bacteria such as Akkermansia and Clostridium XIVa, known as SCFAs producers [53, 54]. Additionally, the microbiota of centenarians presents an increased abundance of facultative anaerobes such as Escherichia coli and a decrease in Faecalibacterium prausnitzii [68].

Advertisement

3. Dysbiosis, stages of dysbiosis, and disease onset

An individual in a healthy state possesses all bacterial phyla in a specific equilibrium, providing a colonization resistance for exogenous bacteria and preventing the overgrowth of potentially pathogenic bacteria. This state is called eubiosis. Any disruption in this equilibrium is known under the general term of dysbiosis. Dysbiosis occurs due to the loss of beneficial bacteria, which leads to the overgrowth of potentially pathogenic bacteria. The final result of these two evolving stages is the loss of bacterial diversity [69].

3.1 Shannon index and stages of dysbiosis

The Shannon Diversity Index (sometimes called the Shannon–Wiener Index or, erroneously, Shannon-Weaver Index) is used for measuring the diversity of species in a community. The Shannon index considers the percent of each species in an ecosystem [70].

Shannon index is also noted as H is calculated with a certain formula, using the proportion of the whole community, which is made up of species. The H value is proportional to the degree of diversity in a certain community. The index statistically depicts biodiversity regarding abundance, uniformity, and dominance. It can differentiate the degree of variation between populations with the same number of alleles. The loss of rare alternative forms easily influences Shannon index and provides more information than allelic abundance or an ordinary number of alleles [71].

The human gut has over 1000 bacterial species, both beneficial and harmful microorganisms. In physiological conditions, the interactions between gut microbiota and the host can maintain homeostasis and a normal commensal relationship, limiting the possible development of pathogenic bacteria [72].

Dysbiosis appears when homeostasis is altered. In this case, several changes occur: alteration of the bacterial composition of microbiota, modified bacterial metabolic activity, and abnormal distribution of bacteria in the gut. The three stages of dysbiosis are loss of beneficial organisms, excessive growth of potentially harmful microorganisms, and loss of overall microbial diversity. However, these three types usually occur simultaneously. Dysbiosis has several consequences on the host’s general health, even associated with obesity, diabetes, autism, inflammatory bowel disease, autoimmune conditions, neurological disorders, or neoplasia [72].

Levy et al. classified type types of dysbiosis: “bloom of pathobionts, loss of commensals, or loss of diversity” [69], while Vangay et al. described four types (“loss of keystone taxa, loss of diversity, shifts in metabolic capacity, or blooms of pathogens”) [73].

Dysbiosis is influenced by inflammation, infections, diet, xenobiotics, genetics, and environmental factors. An abnormally structured microbiota can affect innate and adaptive gut immunity. These immune pathways affect the colonization niche of gut microbiota by influencing antimicrobial peptides and IgA antibody secretion. Although dysbiosis is associated with many immune-related conditions, it is still needed to be determined if it is, indeed, a cause or a consequence of the disease [69].

3.2 Irritable bowel syndrome

Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder affecting 11% of the global population, and it is defined by recurrent abdominal pain associated with stools of abnormal consistency or frequency, occurring for at least 6 months [74, 75]. According to the Bristol stool form scale, there are four clinical subtypes regarding the bowel habits of the patients: IBS-D (diarrhea-predominant IBS), IBS-C (constipation-predominant IBS), IBS-M (mixed IBS), and IBS-U (unsubtyped IBS) [76, 77].

Although the exact pathogenesis of IBS remains unclear, evidence suggests that gut microbiota could be an important tool in mediating this gastrointestinal disease [76, 78]. Studies showed that patients with IBS can have different bacterial composition and richness of the gut microbiota than healthy individuals. Some possible mechanisms involved in IBS pathogenesis include increased intestinal permeability, changes in the immune system, visceral hypersensitivity, impaired gut motility, emotional distress, genetic alterations [79], and, most recently, gut microbial dysbiosis [80]. Dysbiosis is the term used to describe an unbalanced gut microbiota, frequently discovered in postinfectious IBS [81]. Consequently, dysbiosis can be considered an initiating factor of IBS [81].

The “microbiota–gut–brain” axis theory has been brought forth, confirming the critical part that microbial dysbiosis plays in the emergence of IBS symptoms [78, 82]. According to this theory, environmental variables change the gut microbiota in genetically predisposed people, compromising the integrity of the intestinal epithelial barrier [83]. Following the breach of the intestinal barrier, bacteria engage with the host’s immune system, set off a chain of immunological reactions, and cause low-grade mucosal inflammation in the gut wall [83]. Consequently, changes in sensitivity and motility appear, leading to the onset of IBS symptoms like abdominal pain, bloating, and changes in bowel habits [79].

The latest research by Su et al. on irritable bowel syndrome microbiota alterations revealed new data that could improve the future approach to treating and preventing this gastrointestinal disease [84]. The study showed a significant reduction in bacterial diversity of the gut microbiome in patients with IBS-D and IBS-U but not in those with IBS-C when compared to control groups. IBS-D subjects also suffered a decrease in bacteria richness compared to controls. IBS-D and IBS-U had similar compositional alterations, such as depletion of Firmicutes, Actinobacteriota, Verrucomicrobiota, and Campilobacterota and enrichment of Proteobacteria. Subjects with IBS-C showed different phylum abundance: Verrucomicrobiota and Desulfobacterota as pathogenic bacteria. Escherichia/Shigella were found to increase in all three IBS subtypes, but specifically in IBS-D, where they exhibited the strongest positive correlation with hydrogen sulfide production. Bacterial genera like Sutterella, Faecalibacterium, Bifidobacterium, Prevotella, or Ruminococcus showed decreased levels in all IBS patients [84].

The functional changes described in this study revealed that pathways related to palmitoleate biosynthesis were elevated in IBS-C. In contrast, LACTOSECAT pathway involved in the degradation of lactose and galactose was decreased in IBS-D and IBS-U but increased in IBS-C [84].

The data suggested that IBS patients with depression in all three subtypes had lower levels of Bifidobacterium, Sutterella, Butyricimonas, and Butyricicoccaceae UCG009. Compared to IBS patients without depression, higher levels of Proteus in the gut microbiota analysis and depletion of several pathways involving the production of SCFAs were identified [84]. SCFAs are known for inhibiting inflammatory reactions and malignant growth [80]. A growing body of research supports the relationship between microbial dysbiosis and the emergence of anxiety and depression. For instance, gamma-aminobutyric acid (GABA) is known to be produced by several species of the Lactobacillaceae and Bifidobacteraceae family of bacteria. The central nervous system’s primary inhibitory neurotransmitter, GABA, is crucial in the pathophysiology of mood disorders [85, 86]. Additionally, it has been discovered that bacteria from the genera Bacillus and Escherichia create additional neurotransmitters linked to mood and behavior, including dopamine, serotonin, and norepinephrine [87, 88].

Other studies focused on the importance of the serotonin system as a potential target for IBS treatment [89]. Serotonin synthesis from colonic enterochromaffin cells is encouraged by secondary bile acids and SCFAs, which are mostly generated by Eubacterium, Bacteroides, and Clostridium (clusters IV, XI, XIII, and XIVa) [85]. In addition to its other roles, serotonin is a crucial neurotransmitter that controls gastrointestinal motility [90]. IBS-D patients’ serum serotonin levels were found to be higher, and in IBS-C patients, the serum serotonin levels were lower [91].

The study by Su et al. also confirmed the preexisting data that young age and female gender determine more microbiome alterations in all three IBS subtypes [79, 84]. Nevertheless, the microbiota difference between IBS patients and controls decreased with age in the case of IBS-D and increased in the IBS-C subtype [84].

The dietary factors like alcohol, lactose, whole grain, eggs, red wine, or seafood were considered in the IBS patients microbiota study. The results showed that in all three IBS subtypes, subjects who took lactose daily or regularly (3–5 times per week) exhibited larger microbiome alterations than those who never had lactose intake. In contrast, those who drank red wine achieved the opposite effect [84].

3.3 Inflammatory bowel disease

Inflammatory bowel disease (IBD) is a multifactorial autoimmune intestinal disease comprising Crohn’s disease (CD) and ulcerative colitis (UC) [92]. UC is a long-lasting inflammatory and superficial ulcerative disease of the colon. In contrast, CD is a transmural disease of any part of the gastrointestinal tract, often associated with granuloma formation [92, 93, 94].

InBD has clinical periods of remission and relapse, with its onset at any age, but it is usually diagnosed between 20 and 40 years [95].

InBD’s cause is still unknown, although, in genetically predisposed people, it seems to be maintained by a weakened immune response to gut microbes [95]. This aberrant immunological response is linked to dysregulation of both innate and adaptive immune responses [95]. Specific intestinal epithelial barriers are breached in the case of InBD, and non-resolving mucosal damage is considered a key aspect of the illness [96]. Although the exact etiology is uncertain, it may be linked to an infectious pathogen [97], a chemical substance [98], or a metabolic change most likely caused by diet-mediated dysbiosis [99]. According to recent data, the loss of immunological tolerance caused by gut dysbiosis may cause or exacerbate inflammatory bowel disease [100].

Studies have clarified that the composition of the gut microbiota is different in patients with InBD versus healthy individuals and has its differences in patients with CD versus UC [101, 102]. In the gut microbiota of individuals with UC or CD, the abundance of Roseburia and Phascolarctobacterium was dramatically reduced, but Clostridium levels were elevated [103]. Compared to the InBD groups, the control group had a significantly higher concentration of microorganisms such as Gemmiger formicilis and those from the order Clostridiales [104]. Similarly, Ruminococcaceae family microbial organisms, specifically Ruminococcus species, showed noticeably higher prevalence in controls than in InBD [104]. On the contrary, the gut microbiota from patients with InBD was much more enriched in Blautia producta and Clostridium ramosum than that from the control group [104].

Cohort studies observed that the genus Coprococcus 2, Oxalobacter, and Ruminococcaceae UCG014 were associated with a higher risk of InBD. In contrast, genus Eubacterium ventriosum group and Enterorhabdus were associated with a lower risk of InBD [105]. Genus Coprococcus 2 and the Eubacterium ventriosum group were specifically causally related with UC, Lachnospiraceae UCG001, Ruminococcaceae UCG014, and Oxalobacter were specifically causally connected with CD, according to the analysis from Liu et al. [105]. Similar results have also been reported in mucus samples, where CD patients have a significantly higher abundance of aggressive bacteria, such as the genera Escherichia, Ruminococcus (R. gnavus), and Fusobacteria species, compared to healthy controls, and a significantly lower abundance of bacteria from the genera Faecalibacterium, Coprococcus, and Roseburia [106, 107, 108, 109].

In the past, studies utilizing fecal samples revealed dysbiosis in InBD, which is indicated by an overall reduction in the phylum Firmicutes (for example, Faecalibacterium, Roseburia, and Ruminococcus) and an increase in the phylum Proteobacteria (for example, Enterobacteriaceae) [107, 110, 111, 112]. These alterations in gut microbiota are more frequently encountered in the cases of CD than in those of UC [113].

Various digestive disorders lead to a depletion of F. prausnitzii. However, CD patients exhibit this depletion more frequently [104]. Similar microbial species decrease in CD patients as was confirmed by the investigation of Sankarasubramanian et al. [104]. It did, however, show a divergent abundance in the UC patients. Notably, F. prausnitzii also produces butyrate, an increase of which in UC patients may signify an adaptive advantage, as butyrate is a short-chain fatty acid that is necessary for intestinal epithelial cells to function properly [104].

3.4 Infections or diarrhea associated with antibiotics

Antibiotic administration is a major cause of dysbiosis, particularly a lowered microbial diversity [12, 114, 115].

The resistome is a group of antibiotic resistance genes (ARG) that coexists with the gut microbiome since early childhood [115, 116]. The multidrug-resistant bacteria, which show an increasing prevalence worldwide, are acquiring these ARGs, which helps them adapt to antibiotics [117]. ARGs are responsible for developing multidrug-resistant pathogens, especially after recurrent treatment with antibiotics in the first years of life [116]. Studies nowadays focus on understanding how the resistome interferes with the microbiome in different stages of life or in specific events like acute or chronic infections, as well as on how the quantity, identity, and function of ARGs could provide more perspective into new therapeutic management of the microbiome [115]. Research suggests that human gut resistome tends to enrich after antibiotic consumption. Still, its dimension is already notable even before intake, meaning that a transfer from environmental ARGs to host is happening even before exposure to antibiotic treatments [118].

Antibiotics can interfere with metabolic pathways and can decrease gut microbiome diversity [114, 119]. For instance, anaerobes that produce SCFAs may disappear due to antibiotic-induced alterations in the gut microbiota, which may also disturb the metabolism of carbohydrates and bile and cause an osmotic imbalance [120]. Following antibiotic intake, all three intestinal barriers are affected: the epithelial intestinal cells, the mucus and antimicrobial peptides layer, and the immunoprotective layer composed of different immune cells and various biomolecules. This event can interfere with the production of mucin, cytokines, and antimicrobial peptides, dysregulating intestinal function and leading to other infections or even causing recurrent episodes of infections [116].

Antibiotic-associated diarrhea (AAD) is a condition with a prevalence between 5 and70% in adults caused by overgrowth of pathogenic or opportunistic strains in the gut microbiome following antibiotic intake [116]. These microorganisms have each very different disease manifestations and affect specific mechanisms in the gut, but they are all linked to dysbiosis and structural damage of the intestinal wall [120].

Clostridoides difficile infection is the most common cause of nosocomial antibiotic-associated diarrhea in the adult population [121]. Risk factors include age over 65 years, long hospitalization in intensive care, and administering antibiotics (fluoroquinolones, clindamycin, cephalosporins, and beta-lactams in particular) or proton pump inhibitors [122, 123, 124, 125, 126, 127, 128].

Traveler’s diarrhea (TD) is defined as 3 or more liquid stools over 24 hours in patients traveling and has three degrees of disease severity. Mild TD characterizes as 3 unformed stools without other symptoms and does not interfere with the ability of the travelers to continue their activity. In the case of moderate TD, patients suffer from 3 to 5 or more unformed stools a day, without other symptoms, compared to severe TD that consists of more than 5 stools in 24 hours associated with abdominal cramps, fever, vomiting, tenesmus, or blood in feces [129].

Bacteria, viruses, protozoa, and fungi can all cause TD. In addition, enteroaggregative E. coli, other E. coli pathotypes, noroviruses, rotaviruses, Salmonella spp., Campylobacter jejuni, and Shigella spp. could also be responsible for TD. Recent papers have suggested that enteropathogenic E. coli and enteroaggregative E. coli are more common than enterotoxigenic E. coli—producing heat-labile or heat-stable toxin, which was previously thought to be the most frequent cause of TD [129].

Studies regarding the alterations in the diversity and composition of gut microbiota after antibiotic consumption confirm that F/B radio and Enterococcus levels increase [130]. Consequently, diet and food supplements like probiotics or prebiotics can improve gut health by restoring the balance between the species forming the intestinal microbiome [131].

3.5 Colon cancer

Colorectal cancer (CRC) is one of the most frequent cancers, and it carries a major health burden around the globe. It is the third most frequent cancer worldwide and the second in mortality among all cancers [76]. As it is with the majority of cancers, tumor formation in the colon depends on both genetic and environmental factors. Recent studies have suggested that the microbiota is an important environmental factor for some cancers, and it might play an important role in oncogenesis [132].

The theory that microorganisms might be involved in carcinogenesis has been demonstrated as early as the early sixties, using carcinogens in germ-free and conventional mice [133]. Human studies have shown that dysbiosis is also present in the microbiota of CRC patients and that there are metagenomic differences between the gut microbiota of patients diagnosed with CRC and that of healthy subjects. These studies have shown that compared to controls, the microbiota of CRC patients presented reduced microbial diversity, a higher richness in genes, increased amounts of procarcinogenic bacterial taxa (Bacteroides, Escherichia, Fusobacterium, and Porphyromonas) and reduced numbers of health-promoting taxa (e.g., Roseburia) [134, 135, 136, 137]. Apart from Fusobacterium nucleatum, whose connection with CRC has been extensively described, studies have identified other strains of bacteria to be enriched in CRC microbiota, including Porphyromonas asaccharolytica, Poststreprococcus stomatis, Parvimonas micra, and Solobacterium moorei [134, 137, 138, 139, 140, 141, 142, 143]. Other specific bacteria, such as Bacteroides fragilis, E. coli, Enterococcus faecalis, and Streptococcus gallolyticus, have also been associated with CRC in several studies [144, 145, 146].

Conversely, several health-promoting bacteria appear to be depleted in CRC microbiota, such as Streptococcus termophilus and the butyrate-producing genus Roseburia and other butyrate-producing bacteria like Lachnospiraceae and Ruminococcaceae [147, 148, 149, 150].

It appears that certain bacteria not only are pro-oncogenic but also have the capacity to remodel the gut microbiota to promote CRC progression [140].

The mechanisms by which these pro-oncogenic bacteria promote tumorigenesis and cancer progression include the production of genotoxins, inflammation, and immune regulation [147]. The relationship between chronic inflammation and cancer has been established and intensively described. Inflammation leads to mutagenesis and accumulation of mutations Tp53 and other genes in the intestinal epithelial cells [151]. Chronic inflammation of the intestinal epithelial cells reduces the barrier function and exposes stem cells to genotoxins [151]. The link between inflammation and cancer is well exemplified by the increased risk patients with InBD have for developing CRC [152]. Interestingly, studies on colitis mouse modes have demonstrated that cancer did not originate in germ-free mice or in mice treated with antibiotics, underlying the microbiota’s important role in colitis-associated cancer [153, 154]. The pro-oncogenic bacterium Fusobacterium nucleatum promotes intestinal tumorigenesis by activating the nuclear factor-κB (NF-κB) pathway and recruiting tumor-infiltrating myeloid cells, as demonstrated in Apc(Min/+) mouse models [155]. Furthermore, enterotoxigenic Bacteroides fragilis secretes Bacteroides fragilis toxin through which it can induce cancer formation in colonic epithelial cells by activating the inflammatory cascade involving IL-17 signal transducer and activator of transcription 3 and NF-κB signaling [150].

Another carcinogenic mechanism that involves the gut microbiota is represented by the production of genotoxins that further damage the DNA and lead to tumor formation. For example, E. coli secretes colibactin via the pks genomic island, a toxin capable of causing DNA damage, inducing genomic alterations and promoting genomic instability [156]. A second genotoxin that can induce DNA damage is the cytolethal distending toxin produced by pathogenic E. coli strains and Campylobacter spp., as studies have demonstrated [157, 158].

The major role the gut microbiota plays in the development and progression of CRC underlines the importance of further studies that focus on modulating the microbiota to prevent CRC and improve treatment outcomes. Secondly, understanding the changes in the CRC microenvironment can help select microbial biomarkers useful in early diagnosis and treatment evaluation.

3.6 Metabolic syndrome

Metabolic syndrome (MetS) has an alarming increasing prevalence around the world. Its complications are cardiovascular and can also concern nonalcoholic fatty liver or obstructive sleep apnea [5]. The pathophysiology behind MetS comprises a perpetual state of chronic inflammation, insulin resistance, autonomic system dysfunction, and oxidative stress [159, 160].

The term was first used in 1970, but in 1988, Gerald Reaven changed the name of MetS to “syndrome X,” adding insulin resistance to the concept [161]. Over the years, several attempts were made to define the MetS. It is now known that it comprises central obesity, hyperglycemia, dyslipidemia, and arterial hypertension, but more attention is lately drawn to gut microbiome [162].

An important, rather newly, investigated risk factor for MetS is the imbalance in the gut microbiota composition, with significant structural differences compared to the healthy subjects. The pathophysiological chain in MetS involves low-grade inflammation, alteration of the intestinal barrier, and production of metabolites that alter the metabolism and hormone synthesis, leading to a continuous cycle that further only favors MetS [5].

In patients with MetS and high levels of gamma-glutamyl transpeptidase (GGT) versus those with only MetS, with normal levels of GGT, the gut microbial composition was significantly different, noting the presence of unfavorable bacteria in higher concentrations (Megamonas hypermegale, Megamonas funiformis, Klebsiella pneumoniae, and Fusobacterium mortiferum) and low concentrations of “good” bacteria, such as Faecalibacterium prausnitzii, Eubacterium eligens, Bifidobacterium longum, Bifidobacterium pseudocatenulatum, Bacteroides dorei, and Alistipes putredinis [163].

3.7 Obesity

Due to the expanding western lifestyle, obesity rates among young individuals are expanding. The gut microbiota of obese individuals is significantly different from that of healthy ones. For instance, a study on animals fed with high-fat diets revealed high levels of Firmicutes, Proteobacteria, and Verrucomicrobia phylum and low levels of Bacteroidetes. It was reported that these changes were reversed after the cessation of a high-fat diet [164].

Overweight subjects possess an augmented F/B ratio when compared to normal-weight subjects. Firmicutes bacteria can digest long-chain carbohydrates, generating significant amounts of nutrients, and higher energy uptake, favoring weight gain and obesity [165, 166]. The microbiota of obese subjects can store more energy from diet compared to normal-weight controls [167].

Low levels of Ruminococcaceae and Clostridia were noted in obese subjects [168, 169]. Consistent data showed a high abundance of Bacilli and its families Streptococcaceae and Lactobacillaceae and diminished accumulations of Clostridia (Christensenellaceae, Clostridiaceae, and Dehalobacteriaceae) [169].

Operational taxonomic units belonging to the Firmicutes phylum had the most significant associations with adipose tissue. Still, one must keep in mind that Firmicutes, however, is the phylum that dominates the human gut. Interestingly, Oscillospora, Lachnospira, and Ruminococcus operational taxonomic units suggested a protective cardiovascular role, negatively associated with visceral fat mass, while Blautia operational taxonomic units proved the opposite [170]. Blautia genus was noticed as a risk factor, being positively associated with visceral fat [168].

A randomized, double-blind, placebo-controlled trial that concerned daily oral administration of Akkermansia muciniphila for 3 months in obese or overweight individuals led to enhanced insulin sensitivity and lower levels of insulin and total plasma cholesterol [171].

In animal subjects with high-fat diets, oligofructose influenced appetite and body weight by controlling the production of gut peptides. Metabolites resulting from fiber fermentation lead to the production of endogenous glucagon-like peptides 1 and 2 (GLP1, GLP2), GLP1 providing beneficial effects on glucose metabolism. At the same time, GLP2 is involved in the stability of epithelial tight junctions [172, 173].

3.8 Diabetes

Either elevated fasting glucose or type 2 diabetes mellitus (T2DM) belongs to MetS. Chronic inflammation is linked to obesity and insulin resistance. However, not only type 2 diabetes but also type 1 diabetes is influenced by the gut microbiome [174].

The major source of energy for the gut microflora is carbohydrates. T2DM patients usually consume less fiber, dietary fiber being associated with high microbial diversity [175]. Fiber consumption is responsible for a stable microflora [176], favors the fermentation of gut microbes, and amplifies the production of SCFAs. This is essential in host metabolism since SCFAs are in glucose homeostasis [177].

The gut microbiota of subjects with T2DM is characterized by a certain degree of dysbiosis [5]. The gut microbiota in T2DM is mainly composed of Bacteroides, Faecalibacterium, Akkermansia, Ruminococcus, and Fusobacterium, with lower levels of Roseburia [178].

At the debut of T2DM, some evidence suggests the rich presence of Bacteroidetes and diminished populations of Firmicutes [179].

In T2DM patients, gut microbiota is significantly lower in Firmicutes, but nevertheless, it is rich in Gram-negative bacteria belonging to Proteobacteria and Bacteroidetes [180]. The Bacteroidetes/Firmicutes and Bacteroides-Prevotella/C. coccoides-Eubacterium rectale ratios were higher in T2DM patients and positively correlated with glycemic levels [180, 181]. Excessive populations of Actinobacteria and Bacteroidetes phyla can be found in T2DM subjects [182]. However, evidence suggests that reduced concentrations of Lactobacillaceae are associated with GLP1 resistance [183].

However, it is worth mentioning that low levels of Bifidobacteria and F/B ratio were noted in type 1 diabetes [182]. In T2DM, there are lower levels of butyrate-producing bacteria, such as Bifidobacterium, Akkermansia, and Faecalibacterium and also diminished concentrations of the Firmicutes phylum, Clostridiaceae, and Peptostreptococcaceaea families [182].

Butyrate, a short-chain fatty acid, is an important result of dietary fiber fermentation in the gut. It is energy fuel for intestinal epithelial cells; it can favor satiety, lower inflammation, oncogenesis, and oxidative stress and enhance gut barrier function. Furthermore, butyrate has a role in the defense against obesity and insulin resistance [184]. Butyrate is also important in diabetes since it promotes the normal function of pancreatic beta cells [185].

Higher concentrations of proinflammatory bacteria, such as Escherichia coli, and lower concentrations of antiinflammatory bacteria, such as Faecalibacterium prausnitzii, were noted in T2DM [186, 187].

In T2DM, SCFA production is affected, being influenced by low Faecalibacterium prausnitzii concentrations [187]. Altered production or absorption of propionate was mentioned to elevate the risk for T2DM [188].

Lipopolysaccharides are secreted by Gram-negative gut bacteria, leading to low-grade inflammation by acting on type 4 toll-like receptors, amplifying the likelihood of insulin resistance [189].

3.9 Autoimmune conditions and allergies

An unbalanced immune system, which loses its ability to distinguish self from nonself antigens accurately, is involved in autoimmune diseases. The human gut microbiota plays an essential role since it can maintain the homeostasis of the immune system. It is involved in gut functional integrity, barrier stability, and permeability, determining immune response and the progression of inflammatory conditions [190].

Autoimmune conditions involve excessive or improper production of autoantibodies. Immunity can be altered by either genetic or environmental factors. There is growing evidence that gut microbiota can be applied in autoimmune diseases. The modulations of host immune response through antigen-presenting cells may bring forth antigen presentation and cytokine secretion, influencing the differentiation of T cells and leading to an altered balance between T helper 17 and T regulatory cells. As a result, self and nonself antigen similarity and activated pathogen-derived autoreactive T and B cells generate and promote autoimmunity. Moreover, the gut permeability is modified since the expression of tight junction proteins is altered. Weakened gut barrier integrity is important in the pathophysiology of autoimmunity because this way, the immune system interacts with commensal intestinal bacteria [190].

T helper 17 cell activation can be determined by gut microbiota, such as Enterococcus gallinarum and Bifidobacterium adolescentis [191, 192, 193].

Unfortunately, the incidence of allergic reactions has continually expanded over the past years, especially in developed countries. The pathogenesis is complex, including genetic, epigenetic, and environmental factors. Recently, gut microbiota has become an interesting topic in these afflictions [194].

The most common allergens are represented by pollens, dust, insect bites, foods, and drugs resulting in rhinitis, eczema, asthma, or even more severe reactions leading to anaphylactic shock [195].

The gut microbiota plays a major role in immunity, regulating the tendencies of immune reactions, type 2 immunity, and basophil hematopoiesis and preserving the gut barrier function [196].

Neutral bacteria can transform from probiotics to pathogenic bacteria, influenced by the environment or the health status [196].

SCFAs can modulate local gut FOXP3+ regulatory T cells, influencing immune homeostasis [197].

Food allergies are debated worldwide, with the exact cause still uncertain. There is no clear bacterial profile linked to food allergies. However, some evidence suggests that milk allergy in children with abundant Clostridia and Firmicutes after birth disappeared after some time [198].

The gut microbiota of children allergic to milk was rich in Lachnospiraceae and Ruminococcaceae, according to Canani et al. [199]. In young children with food sensitization to at least one nutriment, Haemophilus, Dialister, Clostridium, and Dorea genera were diminished. In contrast, Citrobacter, Oscillospira, Lactococcus, and Dorea genera were also reduced but strongly associated with food allergies [200].

In egg allergy kids, there was a decrease in Leuconostocaceae families and an increase in Streptococcaceae and Lachnospiraceae genera [199, 201].

Respiratory allergies (allergic rhinitis, and asthma) can also be connected to gut dysbiosis [199].

It was also underlined that dietary fiber intake amplified the concentrations of Bacteroidetes and Actinobacteria and lowered Firmicutes and Proteobacteria, diminishing allergic symptoms in allergic rhinitis [202].

Ege and collaborators evaluated almost 500 children in rural and urban Germany, indicating that concentrations of Acinetobacter, Lactobacillus, and Staphylococcus were inversely correlated with asthma and hay fever [203].

Administration of Bifidobacterium longum IM55 and Lactobacillus plantarum IM76 in animal subjects lowered seric IgE concentrations and interleukin 4 and 5 in the upper respiratory tract [204].

Bifidobacterium breve and nondigestible oligosaccharides inhibited inflammation in the lower respiratory tract of murine models by influencing T cell response [205].

In allergic asthmatic children, low Faecalibacterium prausnitzii and Akkermansia muciniphila were observed. It was suggested that these bacteria could promote the production of anti-inflammatory cytokines (interleukin 10) and prevent the release of proinflammatory cytokines (interleukin 12), thus inhibiting inflammation [205].

Atopic dermatitis is an inflammatory condition characterized by pruritus and eczematous lesions, estimating people of all ages, although predominantly children. The scarcity of Bifidobacterium, Akkermansia, and Faecalibacterium was noted in kids prone to asthma and atopic dermatitis [206].

The gut microbiome of children with atopic dermatitis has an inversely correlated diversity with butyrate-producing bacteria concentrations. It is also characterized by an abundance of Clostridia and low levels of Coprococcus eutactus (butyrate producer) [207].

In what concerns nickel allergy, the administration of probiotic Lactobacillus reuteri strains diminished gastrointestinal and cutaneous manifestations [208]. At the same time, another study pointed out the improvement of nickel sensitivity and ameliorated gut dysbiosis after Lactobacilli or Bifidobacteria administration [209].

Advertisement

4. Diet and environment in modulating the intestinal microbiome

To better understand the systemic connections linking the microbiota to human health, many researchers have resorted to studying the impact of dietary components on the microbiota [210]. Host nutrition is a recognized predictor of gut microbiota dynamics and function [211].

Different nutritional ingredients can change the human gut microbiota extremely quickly and in various ways, resulting in shifts in the population of microorganisms found in the digestive tract. Diet, including the consumption of prebiotics and probiotics, has a significant impact on both the gut microbiome’s composition and the intestinal barrier’s function, as noted in the review by Tamang et al. [212]. Probiotics are defined as live microorganisms that, when administered in adequate amounts, will confer a health benefit on the host (according to WHO). At the same time, prebiotics represent a group of biological nutrients that can be processed by the gastrointestinal microorganisms to produce beneficial SCFAs [213, 214].

Several studies compared the Mediterranean diet (MD) to the high-fat Western diet (WD) and found, firstly, that subjects who ate an MD, rich in healthy bioactive compounds, polysaccharides and polyphenols, had a higher gut microbiota diversity than those who ate a WD and, secondly, that more SCFAs are produced in the case of MD, while WD subjects showed increased risk of metabolic disease, such as obesity and hyperlipidemia [215, 216, 217, 218, 219, 220, 221, 222].

Studies suggest that a prolonged high-fat diet can increase the abundance of Ruminococcus, Dorea, Coprococcus, and Adercreutzia and decrease Anaeroplase and Turicibacter. These changes in the gut microbiota were also associated with obesity and insulin resistance [223].

Polyphenols found in tea leaves showed great results in inhibiting the growth of harmful bacteria in the gut, like Enterobacteriaceae or Bilophila, while enhancing the number of Bacteroides and Bifidobacterium and also promoting the production of SCFAs, which are beneficial for health [224, 225, 226].

Other dietary compounds, like polysaccharides, found, for instance, in the purple potato, can modulate the gut microbiota by increasing the population of Oscillospira and Lachnospiraceae while decreasing Suttella, a harmful bacteria [227]. The microbiota has carbohydrate-active enzymes that can digest polysaccharides, use their metabolites for energy production, and maintain health [228, 229].

Dietary carotenoids administration can modulate the gut microbiota by inhibiting the growth of harmful bacteria like Proteobacteria, Dialister, or Enterobacter and increasing the abundance of Bifidobacterium and, in high doses, raising the number of Lactobacillus strains [230]. Carotenoids are natural pigments that can be found in fruits and vegetables, but they can also be consumed in food or beverages colored with the help of carotenoids or even just as supplements [230, 231].

Nevertheless, microbiome-targeted nutritional interventions with measurable impacts on the microbiome remain elusive due to the microbiome’s highly individualized character and diet [212, 232].

As past studies have shown, physical exercise can also interfere with gut microbiome diversity. Sustaining physical activity determined the increase in Bacteroidetes and a decrease in Firmicutes in the intestinal microbiome, as well as promoted the production of SCFAs, which further enhance the body’s metabolism [233, 234, 235, 236].

Other studies suggested that sedentary individuals have a reduced gut microbiota diversity, which can lead to a wide range of health problems [237, 238]. These conclusions offer new approaches to managing and preventing metabolic disorders, stating that early planning of a proper diet and moderate exercise can reduce the burden on the health system [239].

Advertisement

5. Future perspectives

The hope for the future is to reach a time when personalized microbiome medicine can change how we treat specific diseases, when targeted agents can be used against particular microbiome constituents, with minimal side effects and great results (Figure 2) [115].

Figure 2.

Modulation of gut microbiota. Interventions are supposed to be a combination of dietary lifestyle and additional therapeutic measures. Created with Biorender.com.

The genetic manipulation of barrier protein function in animal models through dietary interventions, including the consumption of probiotics, to establish the causal association between gut microbiome changes and epithelial permeability is a provocative but promising subject [212].

The genetic manipulation of gut bacteria can facilitate a deeper molecular understanding of the interrelation between the microbiome and the host. It can even broaden the treatment options for metabolism alterations and associated conditions [240, 241].

Gut microbiota can serve as a new treatment target for metabolic syndrome. Further research is needed to enforce the necessary measures for the treatment and even prevention of one of the increasingly frequent pathologies of the century.

Advertisement

6. Summary

The authors’ goal in this chapter is to highlight the many methods for modifying gut microbiota in relation to various illnesses. The gut microbiota’s function in the human body is explained in great detail, along with the phyla that make up this “superorganism” as a whole. The unique changes the gut microbiota experiences throughout life, from infancy to adulthood and beyond in elderly adults, are shown in a timeline. This page focuses on dysbiosis, which is the dysregulation of the diversity of species and the proliferation of harmful microorganisms. Studies on various diseases have looked at the function of dysbiosis and its consequences on the development of pathogenic conditions. Understanding the stages of dysbiosis development in the body is made easier by the Shannon index’s explanation and its role in the variety of the microbiota population. In relation to the development and therapy of gut dysbiosis, diseases including irritable bowel syndrome, inflammatory bowel syndrome, colon cancer, infections, or diarrhea linked to antibiotics are examined. Obesity, autoimmune diseases, allergies, and metabolic syndrome are a few more illnesses that offer insight into the systemic effects of gut microbiota imbalance in the body. The authors also concentrated on disseminating knowledge regarding dietary interventions that can control the microbiota, a topic that is very hot and constantly evolving in this particular field. Environmental variables were also considered as potential contributors to the changes that may harm the microbiome.

The importance of a C-section versus a regular delivery as a factor in altering the composition of the gut microbiota was covered by the authors in the second chapter. The infant’s diet, including whether or not it was breastfed, is another significant element that is taken into account quickly. The pertinent findings were made when babies who stopped being breastfed had a microbiota that was similar to that of adults. Preterm or C-section babies were discovered to have more pathogenic bacteria in their guts and to be more prone to gastrointestinal infections or inflammatory illnesses in the future. Later, up until the age of three, babies have a tendency to acquire a gut microbe composition that is more stable and similar to that of an adult, being similar. However, a number of circumstances, like exposure to antibiotics, having pets around, or growing up with siblings, influence this process. The authors then went on to discuss the various theories surrounding a healthy adult gut microbiome, bringing to light some assessment tools, like diversity, which are directly tied to the microbiota’s state of health. The differences in the geriatric microbiota are another topic covered in the second chapter of this publication. The capacity of adaptation and evolution of the intestinal microbiota throughout life is emphasized, the microbial diversity decreasing with advancing age and changing dietary habits.

The third chapter examines dysbiosis from two angles: as a process that is explained in all of its stages and defined using the Shannon Index and as a contributor to numerous illnesses. The first pathology discussed that is thought to be influenced by dysbiosis is irritable bowel syndrome (IBS). The notion of the microbiota–gut–brain axis was highlighted as a crucial element in the emergence of microbial dysbiosis. In this thorough assessment of the literature, specific information about microbial changes in the gut flora as well as IBS symptoms were presented. In order to discover the impact on serotonin metabolism, specific focus was given to the changes in microbiota diversity for persons with IBS because psychological alterations like anxiety or depression were associated with the condition. Both ulcerative colitis and Crohn disease were taken into account while discussing inflammatory bowel illnesses in an effort to establish connections between dysregulations of the gut microbiota and particular strains of bacteria that are predominant in each of the cases in question. Another repercussion of dysbiosis is diarrhea brought on by antibiotic treatment. Researchers are accusing the mechanism through which antibiotics influence the host as the origin of microbial dysregulation. Additionally, several types of diarrheas are mentioned, including infection with Clostridium difficile and traveler’s diarrhea, both of which are distinct conditions than antibiotic-related diarrhea but are intricately related to one another. The authors emphasize that numerous bacteria can be pro-oncogenic and contribute to the development of the disease when they proliferate to abnormal levels that favor tumor genesis as they move on in their discussion of colorectal cancer from the point of dysbiosis. Understanding the changes in the gut microbiota of patients with colorectal cancer may result in groundbreaking findings in the area of the microbial-level therapeutic options for cancer that may improve classic therapies. The researchers also focused on metabolic conditions including obesity, diabetes, and even metabolic syndrome that might result from dysbiosis. Each pathology’s altered mechanisms or the most significant changes to the gut microbiota composition are described, and the differences between them are outlined. In this chapter, along with autoimmune diseases, allergies are also covered as a topic that is of recent interest. The importance of dietary components or specific diets in altering the gut flora is covered in the fourth chapter. Probiotics are described as a dietary therapeutic agent that significantly reduces dysbiosis. The relationship between exercise and a healthy bacterial population in the human gut was brought up once more at this point. On this note, according to the authors, following intestinal dysbiosis, sedentary people are more prone to infections and metabolic diseases.

The purpose of this review is to highlight the importance of research on the topic of dysbiosis and to provide insight into the mechanisms of several disorders that may be associated with the gut microbiome dysregulation, in addition to highlighting the need for customized microbiome therapeutics in the future.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Acronyms and abbreviations

AAD

antibiotic-associated diarrhea

ARG

antibiotic resistance genes

CD

Chron’s disease

CRC

colorectal cancer

F/B

Firmicutes/Bacteroidetes

GABA

gamma-aminobutyric acid

GGT

gamma-glutamyl transpepidase

GI

gastrointestinal

GLP 1, 2

glucagon-like peptides 1 and 2

IBS

irritable bowel syndrome

InBD

inflammatory bowel disease

MD

Mediterranean diet

MetS

metabolic syndrome

SCFAs

short-chain fatty acids

T2DM

type 2 diabetes mellitus

TD

traveler’s diarrhea

UC

ulcerative colitis

WD

Western diet

References

  1. 1. Berg G, Rybakova D, Fischer D, Cernava T, Verges MC, Charles T, et al. Microbiome definition re-visited: Old concepts and new challenges. Microbiome. 2020;8(1):103. DOI: 10.1186/s40168-020-00875-0
  2. 2. Thursby E, Juge N. Introduction to the human gut microbiota. The Biochemical Journal. 2017;474(11):1823-1836. DOI: 10.1042/bcj20160510
  3. 3. Dieterich W, Schink M, Zopf Y. Microbiota in the gastrointestinal tract. Medical Sciences. 2018;6(4):116. DOI: 10.3390/medsci6040116
  4. 4. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiological Reviews. 2010;90(3):859-904. DOI: 10.1152/physrev.00045.2009
  5. 5. Wang PX, Deng XR, Zhang CH, Yuan HJ. Gut microbiota and metabolic syndrome. Chinese Medical Journal. 2020;133(7):808-816. DOI: 10.1097/CM9.0000000000000696
  6. 6. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019;7(1):14. DOI: 10.3390/microorganisms7010014
  7. 7. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312(5778):1355-1359. DOI: 10.1126/science.1124234
  8. 8. Luckey TD. Introduction to intestinal microecology. The American Journal of Clinical Nutrition. 1972;25(12):1292-1294. DOI: 10.1093/ajcn/25.12.1292
  9. 9. Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915-1920. DOI: 10.1126/science.1104816
  10. 10. Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3(1):4-14. DOI: 10.4161/gmic.19320
  11. 11. O'Toole PW, Jeffery IB. Gut microbiota and aging. Science. 2015;350(6265):1214-1215. DOI: 10.1126/science.aac8469
  12. 12. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449(7164):804-810. DOI: 10.1038/nature06244
  13. 13. Integrative HMPRNC. The Integrative human microbiome project. Nature. 2019;569(7758):641-648. DOI: 10.1038/s41586-019-1238-8
  14. 14. Yassour M, Jason E, Hogstrom LJ, Arthur TD, Tripathi S, Siljander H, et al. Strain-level analysis of mother-to-child bacterial transmission during the first few months of life. Cell Host & Microbe. 2018;24(1):146-54 e4. DOI: 10.1016/j.chom.2018.06.007
  15. 15. Sharon I, Quijada NM, Pasolli E, Fabbrini M, Vitali F, Agamennone V, et al. The core human microbiome: Does it exist and how can we find it? A critical review of the concept. Nutrients. 2022;14(14):2872. DOI: 10.3390/nu14142872
  16. 16. Ihekweazu FD, Versalovic J. Development of the pediatric gut microbiome: Impact on health and disease. The American Journal of the Medical Sciences. 2018;356(5):413-423. DOI: 10.1016/j.amjms.2018.08.005
  17. 17. Jimenez E, Fernandez L, Marin ML, Martin R, Odriozola JM, Nueno-Palop C, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Current Microbiology. 2005;51(4):270-274. DOI: 10.1007/s00284-005-0020-3
  18. 18. Oh KJ, Lee SE, Jung H, Kim G, Romero R, Yoon BH. Detection of ureaplasmas by the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. Journal of Perinatal Medicine. 2010;38(3):261-268. DOI: 10.1515/jpm.2010.040
  19. 19. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Science Translational Medicine. 2014;6(237):237ra65. DOI: 10.1126/scitranslmed.3008599
  20. 20. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Scientific Reports. 2016;6:23129. DOI: 10.1038/srep23129
  21. 21. Nagpal R, Tsuji H, Takahashi T, Kawashima K, Nagata S, Nomoto K, et al. Sensitive quantitative analysis of the meconium bacterial microbiota in healthy term infants born vaginally or by cesarean section. Frontiers in Microbiology. 2016;7:1997. DOI: 10.3389/fmicb.2016.01997
  22. 22. Nagpal R, Kurakawa T, Tsuji H, Takahashi T, Kawashima K, Nagata S, et al. Evolution of gut Bifidobacterium population in healthy Japanese infants over the first three years of life: A quantitative assessment. Scientific Reports. 2017;7(1):10097. DOI: 10.1038/s41598-017-10711-5
  23. 23. Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, et al. Gut dysbiosis following C-section instigates higher colonisation of toxigenic Clostridium perfringens in infants. Beneficial Microbes. 2017;8(3):353-365. DOI: 10.3920/BM2016.0216
  24. 24. Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, et al. Ontogenesis of the gut microbiota composition in healthy, full-term, vaginally born and breast-fed infants over the first 3 years of life: A quantitative bird's-eye view. Frontiers in Microbiology. 2017;8:1388. DOI: 10.3389/fmicb.2017.01388
  25. 25. Leiby JS, McCormick K, Sherrill-Mix S, Clarke EL, Kessler LR, Taylor LJ, et al. Lack of detection of a human placenta microbiome in samples from preterm and term deliveries. Microbiome. 2018;6(1):196. DOI: 10.1186/s40168-018-0575-4
  26. 26. Guittar J, Shade A, Litchman E. Trait-based community assembly and succession of the infant gut microbiome. Nature Communications. 2019;10(1):512. DOI: 10.1038/s41467-019-08377-w
  27. 27. Ronan V, Yeasin R, Claud EC. Childhood development and the microbiome-the intestinal microbiota in maintenance of health and development of disease during childhood development. Gastroenterology. 2021;160(2):495-506. DOI: 10.1053/j.gastro.2020.08.065
  28. 28. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(26):11971-11975. DOI: 10.1073/pnas.1002601107
  29. 29. Yang I, Corwin EJ, Brennan PA, Jordan S, Murphy JR, Dunlop A. The infant microbiome: Implications for infant health and neurocognitive development. Nursing Research. 2016;65(1):76-88. DOI: 10.1097/NNR.0000000000000133
  30. 30. Reyman M, van Houten MA, van Baarle D, Bosch A, Man WH, Chu M, et al. Impact of delivery mode-associated gut microbiota dynamics on health in the first year of life. Nature Communications. 2019;10(1):4997. DOI: 10.1038/s41467-019-13014-7
  31. 31. Rastall RA. Bacteria in the gut: Friends and foes and how to alter the balance. The Journal of Nutrition. 2004;134(8 Suppl.):2022S-2026S. DOI: 10.1093/jn/134.8.2022S
  32. 32. Adlerberth I, Lindberg E, Aberg N, Hesselmar B, Saalman R, Strannegard IL, et al. Reduced enterobacterial and increased staphylococcal colonization of the infantile bowel: An effect of hygienic lifestyle? Pediatric Research. 2006;59(1):96-101. DOI: 10.1203/01.pdr.0000191137.12774.b2
  33. 33. Stokholm J, Thorsen J, Chawes BL, Schjorring S, Krogfelt KA, Bonnelykke K, et al. Cesarean section changes neonatal gut colonization. The Journal of Allergy and Clinical Immunology. 2016;138(3):881-9 e2. DOI: 10.1016/j.jaci.2016.01.028
  34. 34. Mueller NT, Whyatt R, Hoepner L, Oberfield S, Dominguez-Bello MG, Widen EM, et al. Prenatal exposure to antibiotics, cesarean section and risk of childhood obesity. International Journal of Obesity. 2015;39(4):665-670. DOI: 10.1038/ijo.2014.180
  35. 35. Reyman M, van Houten MA, van Baarle D, Bosch A, Man WH, Chu M, et al. Author correction: Impact of delivery mode-associated gut microbiota dynamics on health in the first year of life. Nature Communications. 2019;10(1):5352. DOI: 10.1038/s41467-019-13373-1
  36. 36. Backhed F, Roswall J, Peng Y, Feng Q , Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host & Microbe. 2015;17(6):852. DOI: 10.1016/j.chom.2015.05.012
  37. 37. Jacquot A, Neveu D, Aujoulat F, Mercier G, Marchandin H, Jumas-Bilak E, et al. Dynamics and clinical evolution of bacterial gut microflora in extremely premature patients. The Journal of Pediatrics. 2011;158(3):390-396. DOI: 10.1016/j.jpeds.2010.09.007
  38. 38. Arboleya S, Binetti A, Salazar N, Fernandez N, Solis G, Hernandez-Barranco A, et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiology Ecology. 2012;79(3):763-772. DOI: 10.1111/j.1574-6941.2011.01261.x
  39. 39. Barrett E, Kerr C, Murphy K, O'Sullivan O, Ryan CA, Dempsey EM, et al. The individual-specific and diverse nature of the preterm infant microbiota. Archives of Disease in Childhood. Fetal and Neonatal Edition. 2013;98(4):F334-F340. DOI: 10.1136/archdischild-2012-303035
  40. 40. Claud EC, Walker WA. Hypothesis: Inappropriate colonization of the premature intestine can cause neonatal necrotizing enterocolitis. The FASEB Journal. 2001;15(8):1398-1403. DOI: 10.1096/fj.00-0833hyp
  41. 41. Siggers RH, Siggers J, Thymann T, Boye M, Sangild PT. Nutritional modulation of the gut microbiota and immune system in preterm neonates susceptible to necrotizing enterocolitis. The Journal of Nutritional Biochemistry. 2011;22(6):511-521. DOI: 10.1016/j.jnutbio.2010.08.002
  42. 42. Ventin-Holmberg R, Saqib S, Korpela K, Nikkonen A, Peltola V, Salonen A, et al. The effect of antibiotics on the infant gut fungal microbiota. Journal of Fungi. 2022;8(4):328. DOI: 10.3390/jof8040328
  43. 43. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222-227. DOI: 10.1038/nature11053
  44. 44. Fontana A, Panebianco C, Picchianti-Diamanti A, Lagana B, Cavalieri D, Potenza A, et al. Gut microbiota profiles differ among individuals depending on their region of origin: An Italian pilot study. International Journal of Environmental Research and Public Health. 2019;16(21):4065. DOI: 10.3390/ijerph16214065
  45. 45. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022-1023. DOI: 10.1038/4441022a
  46. 46. Stojanov S, Berlec A, Strukelj B. The influence of probiotics on the Firmicutes/Bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease. Microorganisms. 2020;8(11):1715. DOI: 10.3390/microorganisms8111715
  47. 47. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature. 2011;473(7346):174-180. DOI: 10.1038/nature09944
  48. 48. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar RD. Role of the normal gut microbiota. World Journal of Gastroenterology. 2015;21(29):8787-8803. DOI: 10.3748/wjg.v21.i29.8787
  49. 49. Shanahan F, Ghosh TS, O'Toole PW. The healthy microbiome-what is the definition of a healthy gut microbiome? Gastroenterology. 2021;160(2):483-494. DOI: 10.1053/j.gastro.2020.09.057
  50. 50. Sonnenburg ED, Sonnenburg JL. The ancestral and industrialized gut microbiota and implications for human health. Nature Reviews. Microbiology. 2019;17(6):383-390. DOI: 10.1038/s41579-019-0191-8
  51. 51. Nishijima S, Suda W, Oshima K, Kim SW, Hirose Y, Morita H, et al. The gut microbiome of healthy Japanese and its microbial and functional uniqueness. DNA Research. 2016;23(2):125-133. DOI: 10.1093/dnares/dsw002
  52. 52. Deng F, Li Y, Zhao J. The gut microbiome of healthy long-living people. Aging (Albany NY). 2019;11(2):289-290. DOI: 10.18632/aging.101771
  53. 53. Kong F, Hua Y, Zeng B, Ning R, Li Y, Zhao J. Gut microbiota signatures of longevity. Current Biology. 2016;26(18):R832-R8R3. DOI: 10.1016/j.cub.2016.08.015
  54. 54. Kong F, Deng F, Li Y, Zhao J. Identification of gut microbiome signatures associated with longevity provides a promising modulation target for healthy aging. Gut Microbes. 2019;10(2):210-215. DOI: 10.1080/19490976.2018.1494102
  55. 55. Claesson MJ, Cusack S, O'Sullivan O, Greene-Diniz R, de Weerd H, Flannery E, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(Suppl 1):4586-4591. DOI: 10.1073/pnas.1000097107
  56. 56. Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, et al. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiology. 2016;16:90. DOI: 10.1186/s12866-016-0708-5
  57. 57. Claesson MJ, Jeffery IB, Conde S, Power SE, O'Connor EM, Cusack S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488(7410):178-184. DOI: 10.1038/nature11319
  58. 58. Collino S, Montoliu I, Martin FP, Scherer M, Mari D, Salvioli S, et al. Metabolic signatures of extreme longevity in northern Italian centenarians reveal a complex remodeling of lipids, amino acids, and gut microbiota metabolism. PLoS One. 2013;8(3):e56564. DOI: 10.1371/journal.pone.0056564
  59. 59. Verdi S, Jackson MA, Beaumont M, Bowyer RCE, Bell JT, Spector TD, et al. An investigation into physical frailty as a link between the gut microbiome and cognitive health. Frontiers in Aging Neuroscience. 2018;10:398. DOI: 10.3389/fnagi.2018.00398
  60. 60. He F, Ouwehand AC, Isolauri E, Hosoda M, Benno Y, Salminen S. Differences in composition and mucosal adhesion of bifidobacteria isolated from healthy adults and healthy seniors. Current Microbiology. 2001;43(5):351-354. DOI: 10.1007/s002840010315
  61. 61. Hopkins MJ, Macfarlane GT. Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. Journal of Medical Microbiology. 2002;51(5):448-454. DOI: 10.1099/0022-1317-51-5-448
  62. 62. Makivuokko H, Tiihonen K, Tynkkynen S, Paulin L, Rautonen N. The effect of age and non-steroidal anti-inflammatory drugs on human intestinal microbiota composition. The British Journal of Nutrition. 2010;103(2):227-234. DOI: 10.1017/S0007114509991553
  63. 63. Rea MC, O'Sullivan O, Shanahan F, O'Toole PW, Stanton C, Ross RP, et al. Clostridium difficile carriage in elderly subjects and associated changes in the intestinal microbiota. Journal of Clinical Microbiology. 2012;50(3):867-875. DOI: 10.1128/JCM.05176-11
  64. 64. Rampelli S, Candela M, Turroni S, Biagi E, Collino S, Franceschi C, et al. Functional metagenomic profiling of intestinal microbiome in extreme ageing. Aging (Albany NY). 2013;5(12):902-912. DOI: 10.18632/aging.100623
  65. 65. Biagi E, Candela M, Turroni S, Garagnani P, Franceschi C, Brigidi P. Ageing and gut microbes: Perspectives for health maintenance and longevity. Pharmacological Research. 2013;69(1):11-20. DOI: 10.1016/j.phrs.2012.10.005
  66. 66. Woodmansey EJ, McMurdo ME, Macfarlane GT, Macfarlane S. Comparison of compositions and metabolic activities of fecal microbiotas in young adults and in antibiotic-treated and non-antibiotic-treated elderly subjects. Applied and Environmental Microbiology. 2004;70(10):6113-6122. DOI: 10.1128/AEM.70.10.6113-6122.2004
  67. 67. Bian G, Gloor GB, Gong A, Jia C, Zhang W, Hu J, et al. The gut microbiota of healthy aged chinese is similar to that of the healthy young. mSphere. 2017;2(5):e00327-17. DOI: 10.1128/mSphere.00327-17
  68. 68. Biagi E, Nylund L, Candela M, Ostan R, Bucci L, Pini E, et al. Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS One. 2010;5(5):e10667. DOI: 10.1371/journal.pone.0010667
  69. 69. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nature Reviews. Immunology. 2017;17(4):219-232. DOI: 10.1038/nri.2017.7
  70. 70. Konopinski MK. Shannon diversity index: A call to replace the original Shannon's formula with unbiased estimator in the population genetics studies. PeerJ. 2020;8:e9391. DOI: 10.7717/peerj.9391
  71. 71. Sherwin WB, Chao A, Jost L, Smouse PE. Information theory broadens the spectrum of molecular ecology and evolution. Trends in Ecology & Evolution. 2017;32(12):948-963. DOI: 10.1016/j.tree.2017.09.012
  72. 72. DeGruttola AK, Low D, Mizoguchi A, Mizoguchi E. Current understanding of dysbiosis in disease in human and animal models. Inflammatory Bowel Diseases. 2016;22(5):1137-1150. DOI: 10.1097/MIB.0000000000000750
  73. 73. Vangay P, Ward T, Gerber JS, Knights D. Antibiotics, pediatric dysbiosis, and disease. Cell Host & Microbe. 2015;17(5):553-564. DOI: 10.1016/j.chom.2015.04.006
  74. 74. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: A meta-analysis. Clinical Gastroenterology and Hepatology. 2012;10(7):712-21 e4. DOI: 10.1016/j.cgh.2012.02.029
  75. 75. Mearin F, Lacy BE, Chang L, Chey WD, Lembo AJ, Simren M, et al. Bowel disorders. Gastroenterology. 2016. DOI: 10.1053/j.gastro.2016.02.031
  76. 76. Han L, Zhao L, Zhou Y, Yang C, Xiong T, Lu L, et al. Altered metabolome and microbiome features provide clues in understanding irritable bowel syndrome and depression comorbidity. The ISME Journal. 2022;16(4):983-996. DOI: 10.1038/s41396-021-01123-5
  77. 77. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scandinavian Journal of Gastroenterology. 1997;32(9):920-924. DOI: 10.3109/00365529709011203
  78. 78. Quigley EMM. The gut-brain Axis and the microbiome: Clues to pathophysiology and opportunities for novel management strategies in irritable bowel syndrome (IBS). Journal of Clinical Medicine. 2018;7(1):6. DOI: 10.3390/jcm7010006
  79. 79. Ford AC, Sperber AD, Corsetti M, Camilleri M. Irritable bowel syndrome. Lancet. 2020;396(10263):1675-1688. DOI: 10.1016/S0140-6736(20)31548-8
  80. 80. Mamieva Z, Poluektova E, Svistushkin V, Sobolev V, Shifrin O, Guarner F, et al. Antibiotics, gut microbiota, and irritable bowel syndrome: What are the relations? World Journal of Gastroenterology. 2022;28(12):1204-1219. DOI: 10.3748/wjg.v28.i12.1204
  81. 81. Kim GH, Lee K, Shim JO. Gut bacterial dysbiosis in irritable bowel syndrome: A case-control study and a cross-cohort analysis using publicly available data sets. Microbiology Spectrum. 2023;11(1):e0212522. DOI: 10.1128/spectrum.02125-22
  82. 82. Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nature Reviews. Gastroenterology & Hepatology. 2009;6(5):306-314. DOI: 10.1038/nrgastro.2009.35
  83. 83. Holtmann GJ, Ford AC, Talley NJ. Pathophysiology of irritable bowel syndrome. The Lancet Gastroenterology & Hepatology. 2016;1(2):133-146. DOI: 10.1016/S2468-1253(16)30023-1
  84. 84. Su Q , Tun HM, Liu Q , Yeoh YK, Mak JWY, Chan FK, et al. Gut microbiome signatures reflect different subtypes of irritable bowel syndrome. Gut Microbes. 2023;15(1):2157697. DOI: 10.1080/19490976.2022.2157697
  85. 85. Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Research. 2018;1693(Pt B):128-133. DOI: 10.1016/j.brainres.2018.03.015
  86. 86. Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. Psychobiotics and the manipulation of bacteria-gut-brain signals. Trends in Neurosciences. 2016;39(11):763-781. DOI: 10.1016/j.tins.2016.09.002
  87. 87. Baj A, Moro E, Bistoletti M, Orlandi V, Crema F, Giaroni C. Glutamatergic signaling along the microbiota-gut-brain axis. International Journal of Molecular Sciences. 2019;20(6):1482. DOI: 10.3390/ijms20061482
  88. 88. Kawase T, Nagasawa M, Ikeda H, Yasuo S, Koga Y, Furuse M. Gut microbiota of mice putatively modifies amino acid metabolism in the host brain. The British Journal of Nutrition. 2017;117(6):775-783. DOI: 10.1017/S0007114517000678
  89. 89. Adak A, Khan MR. An insight into gut microbiota and its functionalities. Cellular and Molecular Life Sciences. 2019;76(3):473-493. DOI: 10.1007/s00018-018-2943-4
  90. 90. Ge X, Pan J, Liu Y, Wang H, Zhou W, Wang X. Intestinal crosstalk between microbiota and serotonin and its impact on gut motility. Current Pharmaceutical Biotechnology. 2018;19(3):190-195. DOI: 10.2174/1389201019666180528094202
  91. 91. Camilleri M, Ford AC. Irritable bowel syndrome: Pathophysiology and current therapeutic approaches. Handbook of Experimental Pharmacology. 2017;239:75-113. DOI: 10.1007/164_2016_102
  92. 92. Louis E, Van Kemseke C, Reenaers C. Necessity of phenotypic classification of inflammatory bowel disease. Best Practice & Research. Clinical Gastroenterology. 2011;25(Suppl 1):S2-S7. DOI: 10.1016/S1521-6918(11)70003-8
  93. 93. Levine A, Griffiths A, Markowitz J, Wilson DC, Turner D, Russell RK, et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: The Paris classification. Inflammatory Bowel Diseases. 2011;17(6):1314-1321. DOI: 10.1002/ibd.21493
  94. 94. Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal classification of inflammatory bowel disease: Controversies, consensus, and implications. Gut. 2006;55(6):749-753. DOI: 10.1136/gut.2005.082909
  95. 95. Saez A, Herrero-Fernandez B, Gomez-Bris R, Sanchez-Martinez H, Gonzalez-Granado JM. Pathophysiology of inflammatory bowel disease: Innate immune system. International Journal of Molecular Sciences. 2023;24(2):1526. DOI: 10.3390/ijms24021526
  96. 96. Leppkes M, Neurath MF. Cytokines in inflammatory bowel diseases - Update 2020. Pharmacological Research. 2020;158:104835. DOI: 10.1016/j.phrs.2020.104835
  97. 97. Mann EA, Saeed SA. Gastrointestinal infection as a trigger for inflammatory bowel disease. Current Opinion in Gastroenterology. 2012;28(1):24-29. DOI: 10.1097/MOG.0b013e32834c453e
  98. 98. Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, et al. Environmental triggers in IBD: A review of progress and evidence. Nature Reviews. Gastroenterology & Hepatology. 2018;15(1):39-49. DOI: 10.1038/nrgastro.2017.136
  99. 99. Schroeder BO, Birchenough GMH, Stahlman M, Arike L, Johansson MEV, Hansson GC, et al. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host & Microbe. 2018;23(1):27-40 e7. DOI: 10.1016/j.chom.2017.11.004
  100. 100. Fava F, Danese S. Intestinal microbiota in inflammatory bowel disease: Friend of foe? World Journal of Gastroenterology. 2011;17(5):557-566. DOI: 10.3748/wjg.v17.i5.557
  101. 101. Glassner KL, Abraham BP, Quigley EMM. The microbiome and inflammatory bowel disease. The Journal of Allergy and Clinical Immunology. 2020;145(1):16-27. DOI: 10.1016/j.jaci.2019.11.003
  102. 102. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59-65. DOI: 10.1038/nature08821
  103. 103. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biology. 2012;13(9):R79. DOI: 10.1186/gb-2012-13-9-r79
  104. 104. Sankarasubramanian J, Ahmad R, Avuthu N, Singh AB, Guda C. Gut microbiota and metabolic specificity in ulcerative colitis and Crohn's disease. Frontiers in Medicine. 2020;7:606298. DOI: 10.3389/fmed.2020.606298
  105. 105. Liu B, Ye D, Yang H, Song J, Sun X, Mao Y, et al. Two-sample mendelian randomization analysis investigates causal associations between gut microbial genera and inflammatory bowel disease, and specificity causal associations in ulcerative colitis or Crohn's disease. Frontiers in Immunology. 2022;13:921546. DOI: 10.3389/fimmu.2022.921546
  106. 106. Sheehan D, Moran C, Shanahan F. The microbiota in inflammatory bowel disease. Journal of Gastroenterology. 2015;50(5):495-507. DOI: 10.1007/s00535-015-1064-1
  107. 107. Nishino K, Nishida A, Inoue R, Kawada Y, Ohno M, Sakai S, et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. Journal of Gastroenterology. 2018;53(1):95-106. DOI: 10.1007/s00535-017-1384-4
  108. 108. Becker C, Neurath MF, Wirtz S. The intestinal microbiota in inflammatory bowel disease. ILAR Journal. 2015;56(2):192-204. DOI: 10.1093/ilar/ilv030
  109. 109. Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology. 2017;152(2):327-39 e4. DOI: 10.1053/j.gastro.2016.10.012
  110. 110. Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology. 2008;134(2):577-594. DOI: 10.1053/j.gastro.2007.11.059
  111. 111. Frank DN, Robertson CE, Hamm CM, Kpadeh Z, Zhang T, Chen H, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflammatory Bowel Diseases. 2011;17(1):179-184. DOI: 10.1002/ibd.21339
  112. 112. Nagalingam NA, Lynch SV. Role of the microbiota in inflammatory bowel diseases. Inflammatory Bowel Diseases. 2012;18(5):968-984. DOI: 10.1002/ibd.21866
  113. 113. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, Zheng Z, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139(6):1844-54 e1. DOI: 10.1053/j.gastro.2010.08.049
  114. 114. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biology. 2008;6(11):e280. DOI: 10.1371/journal.pbio.0060280
  115. 115. Schwartz DJ, Langdon AE, Dantas G. Understanding the impact of antibiotic perturbation on the human microbiome. Genome Medicine. 2020;12(1):82. DOI: 10.1186/s13073-020-00782-x
  116. 116. Kesavelu D, Jog P. Current understanding of antibiotic-associated dysbiosis and approaches for its management. Therapeutic Advances in Infectious Disease. 2023;10:20499361231154443. DOI: 10.1177/20499361231154443
  117. 117. Stalder T, Press MO, Sullivan S, Liachko I, Top EM. Linking the resistome and plasmidome to the microbiome. The ISME Journal. 2019;13(10):2437-2446. DOI: 10.1038/s41396-019-0446-4
  118. 118. Crofts TS, Gasparrini AJ, Dantas G. Next-generation approaches to understand and combat the antibiotic resistome. Nature Reviews. Microbiology. 2017;15(7):422-434. DOI: 10.1038/nrmicro.2017.28
  119. 119. Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, et al. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nature Communications. 2018;9(1):2872. DOI: 10.1038/s41467-018-05336-9
  120. 120. Young VB, Schmidt TM. Antibiotic-associated diarrhea accompanied by large-scale alterations in the composition of the fecal microbiota. Journal of Clinical Microbiology. 2004;42(3):1203-1206. DOI: 10.1128/JCM.42.3.1203-1206.2004
  121. 121. Chandrasekaran R, Lacy DB. The role of toxins in Clostridium difficile infection. FEMS Microbiology Reviews. 2017;41(6):723-750. DOI: 10.1093/femsre/fux048
  122. 122. Samarkos M, Mastrogianni E, Kampouropoulou O. The role of gut microbiota in Clostridium difficile infection. European Journal of Internal Medicine. 2018;50:28-32. DOI: 10.1016/j.ejim.2018.02.006
  123. 123. Rohana H, Azrad M, Nitzan O, Adler A, Binyamin D, Koren O, et al. Characterization of Clostridioides difficile strains, the disease severity, and the microbial changes they induce. Journal of Clinical Medicine. 2020;9(12):4099. DOI: 10.3390/jcm9124099
  124. 124. Bartlett JG. Clinical practice. Antibiotic-associated diarrhea. New England Journal of Medicine. 2002;346(5):334-339. DOI: 10.1056/NEJMcp011603
  125. 125. Bignardi GE. Risk factors for Clostridium difficile infection. The Journal of Hospital Infection. 1998;40(1):1-15. DOI: 10.1016/s0195-6701(98)90019-6
  126. 126. Gerding DN, Johnson S, Peterson LR, Mulligan ME, Silva J Jr. Clostridium difficile-associated diarrhea and colitis. Infection Control and Hospital Epidemiology. 1995;16(8):459-477. DOI: 10.1086/648363
  127. 127. Bartlett JG, Gerding DN. Clinical recognition and diagnosis of Clostridium difficile infection. Clinical Infectious Diseases. 2008;46(Suppl 1):S12-S18. DOI: 10.1086/521863
  128. 128. Davies K, Lawrence J, Berry C, Davis G, Yu H, Cai B, et al. Risk factors for primary Clostridium difficile infection; results from the observational study of risk factors for Clostridium difficile infection in hospitalized patients with infective diarrhea (ORCHID). Frontiers in Public Health. 2020;8:293. DOI: 10.3389/fpubh.2020.00293
  129. 129. Danis R, Wawruch M. Travellers' diarrhoea - Prevention, trends and role of microbiome. Central European Journal of Public Health. 2022;30(1):20-25. DOI: 10.21101/cejph.a6740
  130. 130. Zhao L, Zhang Y, Wang Y, Qiao H, Wang Y, Ren J, et al. Gut microbiota diversity of hospitalized older adult patients with and without antibiotic-associated diarrhea. Aging Clinical and Experimental Research. 2023;35(7):1541-1555. DOI: 10.1007/s40520-023-02436-5
  131. 131. Zhang L, Zeng X, Guo D, Zou Y, Gan H, Huang X. Early use of probiotics might prevent antibiotic-associated diarrhea in elderly (>65 years): A systematic review and meta-analysis. BMC Geriatrics. 2022;22(1):562. DOI: 10.1186/s12877-022-03257-3
  132. 132. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nature Medicine. 2019;25(3):377-388. DOI: 10.1038/s41591-019-0377-7
  133. 133. Laqueur GL, McDaniel EG, Matsumoto H. Tumor induction in germfree rats with methylazoxymethanol (MAM) and synthetic MAM acetate. Journal of the National Cancer Institute. 1967;39(2):355-371
  134. 134. Allali I, Delgado S, Marron PI, Astudillo A, Yeh JJ, Ghazal H, et al. Gut microbiome compositional and functional differences between tumor and non-tumor adjacent tissues from cohorts from the US and Spain. Gut Microbes. 2015;6(3):161-172. DOI: 10.1080/19490976.2015.1039223
  135. 135. Feng Q , Liang S, Jia H, Stadlmayr A, Tang L, Lan Z, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nature Communications. 2015;6:6528. DOI: 10.1038/ncomms7528
  136. 136. Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Research. 2012;22(2):299-306. DOI: 10.1101/gr.126516.111
  137. 137. Yu J, Feng Q , Wong SH, Zhang D, Liang QY, Qin Y, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 2017;66(1):70-78. DOI: 10.1136/gutjnl-2015-309800
  138. 138. Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, et al. Human gut microbiome and risk for colorectal cancer. Journal of the National Cancer Institute. 2013;105(24):1907-1911. DOI: 10.1093/jnci/djt300
  139. 139. Zeller G, Tap J, Voigt AY, Sunagawa S, Kultima JR, Costea PI, et al. Potential of fecal microbiota for early-stage detection of colorectal cancer. Molecular Systems Biology. 2014;10(11):766. DOI: 10.15252/msb.20145645
  140. 140. Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS One. 2012;7(6):e39743. DOI: 10.1371/journal.pone.0039743
  141. 141. Thomas AM, Manghi P, Asnicar F, Pasolli E, Armanini F, Zolfo M, et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nature Medicine. 2019;25(4):667-678. DOI: 10.1038/s41591-019-0405-7
  142. 142. Wirbel J, Pyl PT, Kartal E, Zych K, Kashani A, Milanese A, et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nature Medicine. 2019;25(4):679-689. DOI: 10.1038/s41591-019-0406-6
  143. 143. Yachida S, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nature Medicine. 2019;25(6):968-976. DOI: 10.1038/s41591-019-0458-7
  144. 144. Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338(6103):120-123. DOI: 10.1126/science.1224820
  145. 145. Huycke MM, Abrams V, Moore DR. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis. 2002;23(3):529-536. DOI: 10.1093/carcin/23.3.529
  146. 146. Boleij A, Tjalsma H. The itinerary of Streptococcus gallolyticus infection in patients with colonic malignant disease. The Lancet Infectious Diseases. 2013;13(8):719-724. DOI: 10.1016/S1473-3099(13)70107-5
  147. 147. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. The ISME Journal. 2012;6(2):320-329. DOI: 10.1038/ismej.2011.109
  148. 148. Sinha R, Ahn J, Sampson JN, Shi J, Yu G, Xiong X, et al. Fecal microbiota, fecal metabolome, and colorectal cancer interrelations. PLoS One. 2016;11(3):e0152126. DOI: 10.1371/journal.pone.0152126
  149. 149. Warren RL, Freeman DJ, Pleasance S, Watson P, Moore RA, Cochrane K, et al. Co-occurrence of anaerobic bacteria in colorectal carcinomas. Microbiome. 2013;1(1):16. DOI: 10.1186/2049-2618-1-16
  150. 150. Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nature Medicine. 2009;15(9):1016-1022. DOI: 10.1038/nm.2015
  151. 151. Greten FR, Grivennikov SI. Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity. 2019;51(1):27-41. DOI: 10.1016/j.immuni.2019.06.025
  152. 152. Axelrad JE, Lichtiger S, Yajnik V. Inflammatory bowel disease and cancer: The role of inflammation, immunosuppression, and cancer treatment. World Journal of Gastroenterology. 2016;22(20):4794-4801. DOI: 10.3748/wjg.v22.i20.4794
  153. 153. Abreu MT, Peek RM Jr. Gastrointestinal malignancy and the microbiome. Gastroenterology. 2014;146(6):1534-46 e3. DOI: 10.1053/j.gastro.2014.01.001
  154. 154. Irrazabal T, Belcheva A, Girardin SE, Martin A, Philpott DJ. The multifaceted role of the intestinal microbiota in colon cancer. Molecular Cell. 2014;54(2):309-320. DOI: 10.1016/j.molcel.2014.03.039
  155. 155. Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host & Microbe. 2013;14(2):207-215. DOI: 10.1016/j.chom.2013.07.007
  156. 156. Fais T, Delmas J, Barnich N, Bonnet R, Dalmasso G. Colibactin: More than a new bacterial toxin. Toxins (Basel). 2018;10(4):151. DOI: 10.3390/toxins10040151
  157. 157. Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E, Nougayrede JP. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(25):11537-11542. DOI: 10.1073/pnas.1001261107
  158. 158. He Z, Gharaibeh RZ, Newsome RC, Pope JL, Dougherty MW, Tomkovich S, et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut. 2019;68(2):289-300. DOI: 10.1136/gutjnl-2018-317200
  159. 159. Oda E. Historical perspectives of the metabolic syndrome. Clinics in Dermatology. 2018;36(1):3-8. DOI: 10.1016/j.clindermatol.2017.09.002
  160. 160. Battault S, Meziat C, Nascimento A, Braud L, Gayrard S, Legros C, et al. Vascular endothelial function masks increased sympathetic vasopressor activity in rats with metabolic syndrome. American Journal of Physiology. Heart and Circulatory Physiology. 2018;314(3):H497-H507. DOI: 10.1152/ajpheart.00217.2017
  161. 161. Sarafidis PA, Nilsson PM. The metabolic syndrome: A glance at its history. Journal of Hypertension. 2006;24(4):621-626. DOI: 10.1097/01.hjh.0000217840.26971.b6
  162. 162. Dabke K, Hendrick G, Devkota S. The gut microbiome and metabolic syndrome. The Journal of Clinical Investigation. 2019;129(10):4050-4057. DOI: 10.1172/JCI129194
  163. 163. Sheng S, Yan S, Chen J, Zhang Y, Wang Y, Qin Q , et al. Gut microbiome is associated with metabolic syndrome accompanied by elevated gamma-glutamyl transpeptidase in men. Frontiers in Cellular and Infection Microbiology. 2022;12:946757. DOI: 10.3389/fcimb.2022.946757
  164. 164. Tomas J, Mulet C, Saffarian A, Cavin JB, Ducroc R, Regnault B, et al. High-fat diet modifies the PPAR-gamma pathway leading to disruption of microbial and physiological ecosystem in murine small intestine. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(40):E5934-E5E43. DOI: 10.1073/pnas.1612559113
  165. 165. Sanmiguel C, Gupta A, Mayer EA. Gut microbiome and obesity: A plausible explanation for obesity. Current Obesity Reports. 2015;4(2):250-261. DOI: 10.1007/s13679-015-0152-0
  166. 166. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. The American Journal of Clinical Nutrition. 2011;94(1):58-65. DOI: 10.3945/ajcn.110.010132
  167. 167. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031. DOI: 10.1038/nature05414
  168. 168. Le Roy CI, Beaumont M, Jackson MA, Steves CJ, Spector TD, Bell JT. Heritable components of the human fecal microbiome are associated with visceral fat. Gut Microbes. 2018;9(1):61-67. DOI: 10.1080/19490976.2017.1356556
  169. 169. Peters BA, Shapiro JA, Church TR, Miller G, Trinh-Shevrin C, Yuen E, et al. A taxonomic signature of obesity in a large study of American adults. Scientific Reports. 2018;8(1):9749. DOI: 10.1038/s41598-018-28126-1
  170. 170. Beaumont M, Goodrich JK, Jackson MA, Yet I, Davenport ER, Vieira-Silva S, et al. Heritable components of the human fecal microbiome are associated with visceral fat. Genome Biology. 2016;17(1):189. DOI: 10.1186/s13059-016-1052-7
  171. 171. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nature Medicine. 2019;25(7):1096-1103. DOI: 10.1038/s41591-019-0495-2
  172. 172. Cani PD, Neyrinck AM, Maton N, Delzenne NM. Oligofructose promotes satiety in rats fed a high-fat diet: Involvement of glucagon-like Peptide-1. Obesity Research. 2005;13(6):1000-1007. DOI: 10.1038/oby.2005.117
  173. 173. Musso G, Gambino R, Cassader M. Obesity, diabetes, and gut microbiota: The hygiene hypothesis expanded? Diabetes Care. 2010;33(10):2277-2284. DOI: 10.2337/dc10-0556
  174. 174. Gavin PG, Hamilton-Williams EE. The gut microbiota in type 1 diabetes: Friend or foe? Current Opinion in Endocrinology, Diabetes, and Obesity. 2019;26(4):207-212. DOI: 10.1097/MED.0000000000000483
  175. 175. Sircana A, Framarin L, Leone N, Berrutti M, Castellino F, Parente R, et al. Altered gut microbiota in type 2 diabetes: Just a coincidence? Current Diabetes Reports. 2018;18(10):98. DOI: 10.1007/s11892-018-1057-6
  176. 176. Tap J, Furet JP, Bensaada M, Philippe C, Roth H, Rabot S, et al. Gut microbiota richness promotes its stability upon increased dietary fibre intake in healthy adults. Environmental Microbiology. 2015;17(12):4954-4964. DOI: 10.1111/1462-2920.13006
  177. 177. Gholizadeh P, Mahallei M, Pormohammad A, Varshochi M, Ganbarov K, Zeinalzadeh E, et al. Microbial balance in the intestinal microbiota and its association with diabetes, obesity and allergic disease. Microbial Pathogenesis. 2019;127:48-55. DOI: 10.1016/j.micpath.2018.11.031
  178. 178. Li WZ, Stirling K, Yang JJ, Zhang L. Gut microbiota and diabetes: From correlation to causality and mechanism. World Journal of Diabetes. 2020;11(7):293-308. DOI: 10.4239/wjd.v11.i7.293
  179. 179. Chavez-Carbajal A, Pizano-Zarate ML, Hernandez-Quiroz F, Ortiz-Luna GF, Morales-Hernandez RM, De Sales-Millan A, et al. Characterization of the gut microbiota of individuals at different T2D stages reveals a complex relationship with the host. Microorganisms. 2020;8(1):94. DOI: 10.3390/microorganisms8010094
  180. 180. Sedighi M, Razavi S, Navab-Moghadam F, Khamseh ME, Alaei-Shahmiri F, Mehrtash A, et al. Comparison of gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microbial Pathogenesis. 2017;111:362-369. DOI: 10.1016/j.micpath.2017.08.038
  181. 181. Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5(2):e9085. DOI: 10.1371/journal.pone.0009085
  182. 182. Doumatey AP, Adeyemo A, Zhou J, Lei L, Adebamowo SN, Adebamowo C, et al. Gut microbiome profiles are associated with type 2 diabetes in urban Africans. Frontiers in Cellular and Infection Microbiology. 2020;10:63. DOI: 10.3389/fcimb.2020.00063
  183. 183. Grasset E, Puel A, Charpentier J, Collet X, Christensen JE, Terce F, et al. A specific gut microbiota dysbiosis of type 2 diabetic mice induces GLP-1 resistance through an enteric NO-dependent and gut-brain Axis mechanism. Cell Metabolism. 2017;25(5):1075-90 e5. DOI: 10.1016/j.cmet.2017.04.013
  184. 184. Lin HV, Frassetto A, Kowalik EJ Jr, Nawrocki AR, Lu MM, Kosinski JR, et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One. 2012;7(4):e35240. DOI: 10.1371/journal.pone.0035240
  185. 185. Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, et al. Gut microbiota functions: Metabolism of nutrients and other food components. European Journal of Nutrition. 2018;57(1):1-24. DOI: 10.1007/s00394-017-1445-8
  186. 186. Karlsson FH, Tremaroli V, Nookaew I, Bergstrom G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498(7452):99-103. Epub 20130529. DOI: 10.1038/nature12198
  187. 187. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55-60. DOI: 10.1038/nature11450
  188. 188. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vich Vila A, Vosa U, et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nature Genetics. 2019;51(4):600-605. DOI: 10.1038/s41588-019-0350-x
  189. 189. Huang X, Yan D, Xu M, Li F, Ren M, Zhang J, et al. Interactive association of lipopolysaccharide and free fatty acid with the prevalence of type 2 diabetes: A community-based cross-sectional study. Journal of Diabetes Investigation. 2019;10(6):1438-1446. DOI: 10.1111/jdi.13056
  190. 190. Xu H, Liu M, Cao J, Li X, Fan D, Xia Y, et al. The dynamic interplay between the gut microbiota and autoimmune diseases. Journal of Immunology Research. 2019;2019:7546047. DOI: 10.1155/2019/7546047
  191. 191. Manfredo Vieira S, Hiltensperger M, Kumar V, Zegarra-Ruiz D, Dehner C, Khan N, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359(6380):1156-1161. DOI: 10.1126/science.aar7201
  192. 192. Tan TG, Sefik E, Geva-Zatorsky N, Kua L, Naskar D, Teng F, et al. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(50):E8141-E8E50. DOI: 10.1073/pnas.1617460113
  193. 193. Shaheen WA, Quraishi MN, Iqbal TH. Gut microbiome and autoimmune disorders. Clinical and Experimental Immunology. 2022;209(2):161-174. DOI: 10.1093/cei/uxac057
  194. 194. Han P, Gu JQ , Li LS, Wang XY, Wang HT, Wang Y, et al. The association between intestinal bacteria and allergic diseases-cause or consequence? Frontiers in Cellular and Infection Microbiology. 2021;11:650893. DOI: 10.3389/fcimb.2021.650893
  195. 195. Robinson DS, Larche M, Durham SR. Tregs and allergic disease. The Journal of Clinical Investigation. 2004;114(10):1389-1397. DOI: 10.1172/JCI23595
  196. 196. Montecchiani V, Fanos V. Human microbiome and allergy. Pediatric Allergy and Immunology. 2020;31(Suppl. 26):5-7. DOI: 10.1111/pai.13360
  197. 197. Bhaskaran N, Quigley C, Paw C, Butala S, Schneider E, Pandiyan P. Role of short chain fatty acids in controlling T(regs) and immunopathology during mucosal infection. Frontiers in Microbiology. 2018;9:1995. DOI: 10.3389/fmicb.2018.01995
  198. 198. Bunyavanich S, Shen N, Grishin A, Wood R, Burks W, Dawson P, et al. Early-life gut microbiome composition and milk allergy resolution. The Journal of Allergy and Clinical Immunology. 2016;138(4):1122-1130. DOI: 10.1016/j.jaci.2016.03.041
  199. 199. Berni Canani R, Sangwan N, Stefka AT, Nocerino R, Paparo L, Aitoro R, et al. Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. The ISME Journal. 2016;10(3):742-750. DOI: 10.1038/ismej.2015.151
  200. 200. Savage JH, Lee-Sarwar KA, Sordillo J, Bunyavanich S, Zhou Y, O'Connor G, et al. A prospective microbiome-wide association study of food sensitization and food allergy in early childhood. Allergy. 2018;73(1):145-152. DOI: 10.1111/all.13232
  201. 201. Fazlollahi M, Chun Y, Grishin A, Wood RA, Burks AW, Dawson P, et al. Early-life gut microbiome and egg allergy. Allergy. 2018;73(7):1515-1524. DOI: 10.1111/all.13389
  202. 202. Zhang Z, Shi L, Pang W, Liu W, Li J, Wang H, et al. Dietary fiber intake regulates intestinal microflora and inhibits ovalbumin-induced allergic airway inflammation in a mouse model. PLoS One. 2016;11(2):e0147778. DOI: 10.1371/journal.pone.0147778
  203. 203. Ege MJ, Mayer M, Schwaiger K, Mattes J, Pershagen G, van Hage M, et al. Environmental bacteria and childhood asthma. Allergy. 2012;67(12):1565-1571. DOI: 10.1111/all.12028
  204. 204. Seo JH, Kim HY, Jung YH, Lee E, Yang SI, Yu HS, et al. Interactions between innate immunity genes and early-life risk factors in allergic rhinitis. Allergy, Asthma & Immunology Research. 2015;7(3):241-248. DOI: 10.4168/aair.2015.7.3.241
  205. 205. Sagar S, Vos AP, Morgan ME, Garssen J, Georgiou NA, Boon L, et al. The combination of Bifidobacterium breve with non-digestible oligosaccharides suppresses airway inflammation in a murine model for chronic asthma. Biochimica et Biophysica Acta. 2014;1842(4):573-583. DOI: 10.1016/j.bbadis.2014.01.005
  206. 206. Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nature Medicine. 2016;22(10):1187-1191. DOI: 10.1038/nm.4176
  207. 207. Nylund L, Nermes M, Isolauri E, Salminen S, de Vos WM, Satokari R. Severity of atopic disease inversely correlates with intestinal microbiota diversity and butyrate-producing bacteria. Allergy. 2015;70(2):241-244. DOI: 10.1111/all.12549
  208. 208. Randazzo CL, Pino A, Ricciardi L, Romano C, Comito D, Arena E, et al. Probiotic supplementation in systemic nickel allergy syndrome patients: Study of its effects on lactic acid bacteria population and on clinical symptoms. Journal of Applied Microbiology. 2015;118(1):202-211. DOI: 10.1111/jam.12685
  209. 209. Lombardi F, Fiasca F, Minelli M, Maio D, Mattei A, Vergallo I, et al. The effects of Low-nickel diet combined with oral administration of selected probiotics on patients with systemic nickel allergy syndrome (SNAS) and gut dysbiosis. Nutrients. 2020;12(4):1040. DOI: 10.3390/nu12041040
  210. 210. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559-563. DOI: 10.1038/nature12820
  211. 211. Sonnenburg JL, Backhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535(7610):56-64. DOI: 10.1038/nature18846
  212. 212. Lama Tamang R, Juritsch AF, Ahmad R, Salomon JD, Dhawan P, Ramer-Tait AE, et al. The diet-microbiota axis: A key regulator of intestinal permeability in human health and disease. Tissue Barriers. 2023;11(2):2077069. DOI: 10.1080/21688370.2022.2077069
  213. 213. Bamigbade GB, Subhash AJ, Kamal-Eldin A, Nystrom L, Ayyash M. An updated review on prebiotics: Insights on potentials of food seeds waste as source of potential prebiotics. Molecules. 2022;27(18):5947. DOI: 10.3390/molecules27185947
  214. 214. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nature Reviews. Gastroenterology & Hepatology. 2014;11(8):506-514. DOI: 10.1038/nrgastro.2014.66
  215. 215. Shively CA, Appt SE, Vitolins MZ, Uberseder B, Michalson KT, Silverstein-Metzler MG, et al. Mediterranean versus Western diet effects on caloric intake, obesity, metabolism, and hepatosteatosis in nonhuman primates. Obesity (Silver Spring). 2019;27(5):777-784. DOI: 10.1002/oby.22436
  216. 216. Garcia-Montero C, Fraile-Martinez O, Gomez-Lahoz AM, Pekarek L, Castellanos AJ, Noguerales-Fraguas F, et al. Nutritional components in Western diet versus Mediterranean diet at the gut microbiota-immune system interplay. Implications for health and disease. Nutrients. 2021;13(2):699. DOI: 10.3390/nu13020699
  217. 217. Merra G, Noce A, Marrone G, Cintoni M, Tarsitano MG, Capacci A, et al. Influence of Mediterranean diet on human gut microbiota. Nutrients. 2020;13(1):7. DOI: 10.3390/nu13010007
  218. 218. Barber C, Mego M, Sabater C, Vallejo F, Bendezu RA, Masihy M, et al. Differential effects of Western and Mediterranean-type diets on gut microbiota: A metagenomics and metabolomics approach. Nutrients. 2021;13(8):2638. DOI: 10.3390/nu13082638
  219. 219. Liu B, Zhang Y, Wang R, An Y, Gao W, Bai L, et al. Western diet feeding influences gut microbiota profiles in apoE knockout mice. Lipids in Health and Disease. 2018;17(1):159. DOI: 10.1186/s12944-018-0811-8
  220. 220. Bortolin RC, Vargas AR, Gasparotto J, Chaves PR, Schnorr CE, Martinello KB, et al. A new animal diet based on human Western diet is a robust diet-induced obesity model: Comparison to high-fat and cafeteria diets in term of metabolic and gut microbiota disruption. International Journal of Obesity. 2018;42(3):525-534. DOI: 10.1038/ijo.2017.225
  221. 221. Wang Y, You Y, Tian Y, Sun H, Li X, Wang X, et al. Pediococcus pentosaceus PP04 ameliorates high-fat diet-induced hyperlipidemia by regulating lipid metabolism in C57BL/6N mice. Journal of Agricultural and Food Chemistry. 2020;68(51):15154-15163. DOI: 10.1021/acs.jafc.0c05060
  222. 222. Speakman JR. Use of high-fat diets to study rodent obesity as a model of human obesity. International Journal of Obesity. 2019;43(8):1491-1492. DOI: 10.1038/s41366-019-0363-7
  223. 223. Velazquez KT, Enos RT, Bader JE, Sougiannis AT, Carson MS, Chatzistamou I, et al. Prolonged high-fat-diet feeding promotes non-alcoholic fatty liver disease and alters gut microbiota in mice. World Journal of Hepatology. 2019;11(8):619-637. DOI: 10.4254/wjh.v11.i8.619
  224. 224. Chen T, Yang CS. Biological fates of tea polyphenols and their interactions with microbiota in the gastrointestinal tract: Implications on health effects. Critical Reviews in Food Science and Nutrition. 2020;60(16):2691-2709. DOI: 10.1080/10408398.2019.1654430
  225. 225. Liu Z, de Bruijn WJC, Bruins ME, Vincken JP. Reciprocal interactions between Epigallocatechin-3-gallate (EGCG) and human gut microbiota In vitro. Journal of Agricultural and Food Chemistry. 2020;68(36):9804-9815. DOI: 10.1021/acs.jafc.0c03587
  226. 226. Wang L, Zeng B, Liu Z, Liao Z, Zhong Q , Gu L, et al. Green tea polyphenols modulate colonic microbiota diversity and lipid metabolism in high-fat diet treated HFA mice. Journal of Food Science. 2018;83(3):864-873. DOI: 10.1111/1750-3841.14058
  227. 227. Tang C, Sun J, Zhou B, Jin C, Liu J, Kan J, et al. Effects of polysaccharides from purple sweet potatoes on immune response and gut microbiota composition in normal and cyclophosphamide treated mice. Food & Function. 2018;9(2):937-950. DOI: 10.1039/c7fo01302g
  228. 228. Aakko J, Pietila S, Toivonen R, Rokka A, Mokkala K, Laitinen K, et al. A carbohydrate-active enzyme (CAZy) profile links successful metabolic specialization of Prevotella to its abundance in gut microbiota. Scientific Reports. 2020;10(1):12411. DOI: 10.1038/s41598-020-69241-2
  229. 229. Wardman JF, Bains RK, Rahfeld P, Withers SG. Carbohydrate-active enzymes (CAZymes) in the gut microbiome. Nature Reviews. Microbiology. 2022;20(9):542-556. DOI: 10.1038/s41579-022-00712-1
  230. 230. Rocha HR, Coelho MC, Gomes AM, Pintado ME. Carotenoids diet: Digestion, gut microbiota modulation, and inflammatory diseases. Nutrients. 2023;15(10):2265. DOI: 10.3390/nu15102265
  231. 231. Nabi F, Arain MA, Rajput N, Alagawany M, Soomro J, Umer M, et al. Health benefits of carotenoids and potential application in poultry industry: A review. Journal of Animal Physiology and Animal Nutrition. 2020;104(6):1809-1818. DOI: 10.1111/jpn.13375
  232. 232. Johnson AJ, Vangay P, Al-Ghalith GA, Hillmann BM, Ward TL, Shields-Cutler RR, et al. Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host & Microbe. 2019;25(6):789-802 e5. DOI: 10.1016/j.chom.2019.05.005
  233. 233. Denou E, Marcinko K, Surette MG, Steinberg GR, Schertzer JD. High-intensity exercise training increases the diversity and metabolic capacity of the mouse distal gut microbiota during diet-induced obesity. American Journal of Physiology. Endocrinology and Metabolism. 2016;310(11):E982-E993. DOI: 10.1152/ajpendo.00537.2015
  234. 234. Queipo-Ortuno MI, Seoane LM, Murri M, Pardo M, Gomez-Zumaquero JM, Cardona F, et al. Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS One. 2013;8(5):e65465. DOI: 10.1371/journal.pone.0065465
  235. 235. Ortiz-Alvarez L, Xu H, Martinez-Tellez B. Influence of exercise on the human gut microbiota of healthy adults: A systematic review. Clinical and Translational Gastroenterology. 2020;11(2):e00126. DOI: 10.14309/ctg.0000000000000126
  236. 236. Matsumoto M, Inoue R, Tsukahara T, Ushida K, Chiji H, Matsubara N, et al. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Bioscience, Biotechnology, and Biochemistry. 2008;72(2):572-576. DOI: 10.1271/bbb.70474
  237. 237. Castellanos N, Diez GG, Antunez-Almagro C, Bailen M, Bressa C, Gonzalez Soltero R, et al. A critical mutualism - competition interplay underlies the loss of microbial diversity in sedentary lifestyle. Frontiers in Microbiology. 2019;10:3142. DOI: 10.3389/fmicb.2019.03142
  238. 238. Castellanos N, Diez GG, Antunez-Almagro C, Bressa C, Bailen M, Gonzalez-Soltero R, et al. Key bacteria in the gut microbiota network for the transition between sedentary and active lifestyle. Microorganisms. 2020;8:5. DOI: 10.3390/microorganisms8050785
  239. 239. Zhang L, Liu Y, Wang X, Zhang X. Physical exercise and diet: Regulation of gut microbiota to prevent and treat metabolic disorders to maintain health. Nutrients. 2023;15:6. DOI: 10.3390/nu15061539
  240. 240. Chen Z, Jin W, Hoover A, Chao Y, Ma Y. Decoding the microbiome: Advances in genetic manipulation for gut bacteria. Trends in Microbiology. 2023; S0966-842X(23)00160-9. DOI: 10.1016/j.tim.2023.05.007
  241. 241. Jin WB, Li TT, Huo D, Qu S, Li XV, Arifuzzaman M, et al. Genetic manipulation of gut microbes enables single-gene interrogation in a complex microbiome. Cell. 2022;185(3):547-62 e22. DOI: 10.1016/j.cell.2021.12.035

Written By

Bogdan Severus Gaspar, Monica Profir, Oana Alexandra Rosu, Ruxandra Florentina Ionescu and Sanda Maria Cretoiu

Submitted: 22 August 2023 Reviewed: 12 October 2023 Published: 29 May 2024