ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

Gut Microbiome–Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and Disorders

  • Janet M. Sasso
    Janet M. Sasso
    CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
    More by Janet M. Sasso
  • Ramy M. Ammar
    Ramy M. Ammar
    Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
    More by Ramy M. Ammar
  • Rumiana Tenchov
    Rumiana Tenchov
    CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
    More by Rumiana Tenchov
  • Steven Lemmel
    Steven Lemmel
    CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
    More by Steven Lemmel
  • Olaf Kelber
    Olaf Kelber
    Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
    More by Olaf Kelber
  • Malte Grieswelle
    Malte Grieswelle
    Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
  • , and 
  • Qiongqiong Angela Zhou*
    Qiongqiong Angela Zhou
    CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
    *Email: [email protected]
Cite this: ACS Chem. Neurosci. 2023, 14, 10, 1717–1763
Publication Date (Web):May 8, 2023
https://doi.org/10.1021/acschemneuro.3c00127

Copyright © 2023 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY 4.0.
  • Open Access

Article Views

18297

Altmetric

-

Citations

3
LEARN ABOUT THESE METRICS
PDF (5 MB)
Supporting Info (1)»

Abstract

Gut microbiota includes a vast collection of microorganisms residing within the gastrointestinal tract. It is broadly recognized that the gut and brain are in constant bidirectional communication, of which gut microbiota and its metabolic production are a major component, and form the so-called gut microbiome–brain axis. Disturbances of microbiota homeostasis caused by imbalance in their functional composition and metabolic activities, known as dysbiosis, cause dysregulation of these pathways and trigger changes in the blood–brain barrier permeability, thereby causing pathological malfunctions, including neurological and functional gastrointestinal disorders. In turn, the brain can affect the structure and function of gut microbiota through the autonomic nervous system by regulating gut motility, intestinal transit and secretion, and gut permeability. Here, we examine data from the CAS Content Collection, the largest collection of published scientific information, and analyze the publication landscape of recent research. We review the advances in knowledge related to the human gut microbiome, its complexity and functionality, its communication with the central nervous system, and the effect of the gut microbiome–brain axis on mental and gut health. We discuss correlations between gut microbiota composition and various diseases, specifically gastrointestinal and mental disorders. We also explore gut microbiota metabolites with regard to their impact on the brain and gut function and associated diseases. Finally, we assess clinical applications of gut-microbiota-related substances and metabolites with their development pipelines. We hope this review can serve as a useful resource in understanding the current knowledge on this emerging field in an effort to further solving of the remaining challenges and fulfilling its potential.

This publication is licensed under

CC-BY 4.0.
  • cc licence
  • by licence

Introduction

ARTICLE SECTIONS
Jump To

The Earth microbiome represents the majority of the planet’s biodiversity. Microbial life was the first to inhabit Earth. (1) Microbes regulate global nutrient cycles, greenhouse gas exchange, as well as disease transmission and protection, thus providing essential life support to the planet. (2) Among many other harbors, including plants, animals, soil, and entire ecosystems, a wide diversity of microorganisms colonize the human body, which are now known to play an essential role in the human host by regulating key physiological functions.
The large collection of microorganisms inhabiting the human body are predominantly bacteria, but also viruses, protozoa, fungi, and archaea. They are collectively known as the human microbiota. Those microorganisms residing in the digestive tracts are known as gut flora or gut microbiota. As a matter of fact, there are more bacterial cells in the human body than human cells─roughly 40 trillion bacterial cells versus only 30 trillion human cells. Together, they function as an extra organ in the human body─a so-called “forgotten organ”─since these microbes have a collective metabolic activity equal to a virtual organ. (3) The collective genome of the gut microbes, the gut microbiome, exceeds over 100 times the amount of human genome in the body. (4) Considering such enormous genetic potential of the microbiota, it is anticipated that it plays a role in virtually all physiological processes in the human body, including metabolic functions and immune homeostasis. (5−11)
Despite being considered a relatively new field of research, the first reports of human-associated microbiota date back to the 17th century when Antonie van Leeuwenhoek described five different kinds of oral bacteria. (12) In the following decades, the foundations of microbiology were laid, and knowledge of the host–microorganism interactions has accumulated (Figure 1). (13−22) Despite these early findings, rapid development of the field only started when methods to culture anaerobic organisms were set up in the mid-20th century when representatives of the microbiota were grown and studied in the laboratory. (23)

Figure 1

Figure 1. Timeline of major research and development milestones related to the microbiome. (12−30,39,49−80)

In the 2010s, the gut microbiome field burst into the life sciences research and industry, which prompted Forbes to declare the 2010s “The Decade of the Microbiome.” (24) This growth in the field was largely related to the National Institutes of Health’s “The Human Microbiome Project” (25) and the MetaHIT project funded by the European Union. (26) In 2005, the International Human Microbiome Consortium (IHMC) was founded in a cooperative effort to study the microbiome in human health and disease with the ultimate goal of applying this knowledge to prevent and/or treat diseases, and the abovementioned megaprojects have contributed to fulfilling this goal. They provided significant evidence for the relationship between metabolic, neurological, and autoimmune disorders; allergies, infections, and cancers; and the microorganisms that live on and in humans. Specifically, gastrointestinal diseases/disorders, such as inflammatory bowel diseases that include both Crohn’s disease and ulcerative colitis, irritable bowel syndrome (IBS), functional dyspepsia (FD), constipation, celiac disease, and more, are attracting attention with their close relation to the gut microbiome. Because of these findings and the essential part the gut microbiome plays in drug metabolism, the microbiome has become a popular target in the biotechnology industry. In 2010, the first extensive catalogue of human intestinal microbial genes was published on the basis of the studies of 124 individuals. (27) In 2011, the Human Microbiome Project published the sequences of 178 bacterial species. (28)
Although DNA sequencing has been used for decades, it was only after the development of next-generation sequencing when metagenomic studies became affordable. (29) The term metagenomics is used to describe genetic studies of microbial assemblies from environmental samples using sequence-based bioinformatics tools. (30) The goal of these studies is to identify the taxonomic diversity of the microbiota and to differentiate the biological roles of the representatives of such samples by performing functional metagenomics.
The human microbiota plays an essential role in human physiology and pathology. It collaborates closely with the digestive tract in several important aspects: (a) it promotes digestion by assisting the absorption of nutrients by gut cells or the fermentation of some food fractions, which generate important metabolites, including short-chain fatty acids; (31) (b) it supports the maturation of the digestive tract by participating in the assembly of gastrointestinal mucus and promoting the enzymatic activity of the mucosa; (32) (c) it performs a barrier function against pathogens and toxins, where some bacteria release antimicrobial agents that protect from the pathogenic bacteria; (33) (d) it plays a protective role in promoting the immune system development; and (e) it supports in the synthesis of essential vitamins like vitamin B: Magnúsdóttir et al. estimated that 86% of the recommended daily allowance (RDA) of vitamin B6, 37% of the RDA of vitamin B9, 31% of the RDA of vitamin B12, and 27% of the RDA of vitamin B3 could be provided by the human gut microbiota. (34)
Gut microbial disruption (dysbiosis) causes not only gastrointestinal disorders but also disorders in other distal organs and systems. Not long ago, it was found that gut bacteria can affect the central nervous system (CNS) functions. (35−38) Indeed, the gut and brain are in constant bidirectional communication, of which the microbiota and its metabolic production are a major component. The gut and brain connect via a neuro-immuno-humoral network of signaling pathways known as the gut microbiome–brain axis, which includes the vagus nerve, the immune system, the hormonal system, and bacterial metabolites and products. (39) The digestive system, including the inhabiting microbiota, was even called “the second brain” (40) at the time when scientists were beginning to realize that the gut and the brain in humans were involved in constant crosstalk and significantly modulate each other’s function. During disturbance of the microbiota homeostasis caused by an imbalance in their functional composition and metabolic activities, known as dysbiosis, these routes are dysregulated and cause changes in the permeability of the blood–brain barrier (BBB), neuroinflammation, and other pathological malfunctions, including a range of neurodevelopmental and neurodegenerative disorders. (41) Disorders of the gut–brain interaction (DGBI) is the recent term proposed by Rome Foundation guidelines for a range of functional gastrointestinal disorders including but not limited to IBS, FD, and functional constipation. This highlights the central role of the miscommunication between the gut and brain in these digestive disorders. (42) The gut microorganisms transform and metabolize dietary- and host-derived substances to generate a diverse set of metabolites with important local and systemic outcomes, thereby building a network of immunological, neuronal, and endocrine signaling pathways.
It is generally believed that bacterial colonization begins during birth. (43) The neonatal microbiota differs depending on mode of delivery: in vaginally delivered infants, it resembles the maternal vaginal microbiota, while the microbiota in those delivered by cesarean section resembles the maternal skin microbiota. (44) Premature birth, feeding method, and perinatal administration of antibiotics are also among the conditions affecting the development of the neonatal microbiome. (45) Recently, a new mode of horizontal mother-to-infant microbiome transmission has been revealed where microbes in the maternal gut shared genes with microbes in the infant gut during the perinatal period, which starts shortly before birth and prolongs throughout the first few weeks after birth. (46) A major factor of gut microbiota composition during adulthood is diet. Prompt changes in microbiota composition take place in response to dietary style changes. Specific patterns have been reported in plant-based versus animal-based diets. (47,48) The development and modifications of the gut microbiota are influenced by multiple other factors, such as exposure to stress, environmental conditions, medications intake, lifecycle, medical disorders, and procedures.
The human gut microbiota is divided into many groups called phyla. The gut microbiota primarily comprises four main phyla, including Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria, with the Firmicutes and Bacteroidetes representing 90% of gut microbiota. (81,82) The majority of bacteria reside within the gastrointestinal tract, with most predominantly anaerobic bacteria housed in the large intestine. (83)
In recent years, sizable technological progress and wealth of knowledge have promoted the advancement of microbiome research, thereby enhancing our understanding of its relationship to human physiology and pathologies. In this paper, we review the advances in the knowledge related to the human gut microbiome, its complexity and functionality, its communication with the central nervous system, and the effect of the gut microbiome–brain axis on mental and digestive health. We examine data from the CAS Content Collection, (15) the largest human-curated collection of published scientific information and analyze the publication landscape of recent research in order to provide insights into the scientific advances in the area. We also discuss the correlations between the gut microbiota composition and various diseases, specifically digestive system diseases, mental, and neurodegenerative disorders. We furthermore explore the gut microbiota metabolites with regard to their impact on brain, digestive functions, and their associated diseases. Subsequently, we assess the clinical applications of gut microbiota-related substances and metabolites, their development pipelines, disease categories, development stages, and publication trends. We hope this review can serve as a useful resource in understanding the current state of knowledge in the field of gut microbes and the gut microbiome–brain interactions in an effort to further solve the remaining challenges for fulfilling the potential of the field.

Landscape of Gut Microbiome Research─Insights from the CAS Content Collection

ARTICLE SECTIONS
Jump To

The CAS Content Collection (84) is the largest human-curated collection of published scientific knowledge. It is a comprehensive resource to access and remain well-informed on the world’s available scientific literature across disciplines, including chemistry, biomedical sciences, engineering, materials science, agricultural science, and many more, thus allowing quantitative analysis of global scientific publications against variables, such as time, research area, application, disease association, and chemical composition. A search in the CAS Content Collection showed an intense increase of the documents related to microbiome research in the past decade, which overcame other “omics” exploration─for example, the number of proteomics-related documents held up after the initial burst in the early 2000s and were surpassed by the microbiome documents after 2016 (Figure 2, inset). Currently, there are over 250 000 scientific publications (mainly journal articles and patents) in the CAS Content Collection related to gut/intestinal microbiome/microbiota. Nearly 15 000 of them are related to various aspects of mental and gut health. There is a steady, exponential growth of the number of journal articles over time that has been rather explosive from 2021–2022 (Figure 2). The number of patents rapidly grew until 2004, which possibly correlated with the initial accumulation of knowledge and its transfer into patentable applications. Later on, the growth substantially slowed down, perhaps awaiting the forthcoming breakthroughs in the gut microbiome awareness (Figure 2).

Figure 2

Figure 2. Journal and patent publication trends on gut microbiome research related to mental and gut health according to the CAS Content Collection. Inset: microbiome vs proteome document yearly trends.

The United States, China, Japan, and Korea are the leaders in the number of published journal articles (Figure 3A) and patents (Figure 3B) related to gut microbiome research in the areas of mental and gut health.

Figure 3

Figure 3. Top countries publishing journal articles (A) and patents (B) related to gut microbiome research in mental and gut health.

Figure 4 presents the flow of patent filings from different applicant locations to various patent offices. Because patent protection is territorial, and the same invention may be filed for protection in multiple jurisdictions, we looked at all relevant filings on gut microbiome research in mental and gut health. One patent family may have been counted multiple times when it was applied in multiple patent offices. There are diverse patent filing strategies: some patent assignees, such as those from China and Korea, file foremost in their home country patent offices (CN, KR), with a smaller proportion filing through other patent offices or other jurisdictions. Others, for instance United States-based applicants, have a nearly equal number of US and WO filings and a considerable number of filings at other patent offices, such as the European Patent Office (EP) and Canada (CA).

Figure 4

Figure 4. Flow of patent filings related to gut microbiome research in mental and gut health from different patent assignee locations (left) to various patent offices of filing (right). The abbreviations on the right indicate the patent offices of United States (US), Australia (AU), World Intellectual Property Organization (WO), Canada (CA), Austria (AT), European Patent Office (EP), Israel (IL), Norway (NO), Hong Kong (HK), Mexico (MX), Japan (JP), Brazil (BR), Spain (ES), India (IN), Russian Federation (RU), China (CN), and Korea (KR).

In order to better understand the advance in this research area, we examined the occurrence and trends of certain key concepts in the scientific publications relevant to the gut microbiome research in mental and gut health (Figure 5). With respect to the cumulative number of publications, “immunity” and “gut microbiome” appear as top concepts in the area (Figure 5A), thereby reflecting the rising interest in the relationship between the gut microbiome and systemic immune response pathways and the critical role the gut microbiome plays in training and development of the host’s innate and adaptive immune system. It is noteworthy that the concept concerning the gut–brain relationship exhibits the greatest growth rate in the past two years (Figure 5B), thereby characterizing it as the trendiest concept in the field.

Figure 5

Figure 5. Key concepts in the scientific publications relevant to the gut microbiome research in mental and gut health. (A) Number of publications exploring key concepts related to gut microbiome research in mental and gut health. (B) Trends in key concepts presented in the articles related to gut microbiome research in mental and gut health during the years 2016–2021. Percentages are calculated with yearly publication numbers for each key concept, normalized by the total number of publications for the same concept in the same time period.

Gut-Microbiota-Participant Bacteria

ARTICLE SECTIONS
Jump To

The human gut microbiome, as mentioned above, is a complex mixture of microorganisms, including viruses, archaea, bacteria, yeasts, and fungi, interacting with each other and with their host in complex ways. These interactions at various times involve symbiosis, mutualism, antagonism, and even predation. Not only does the gut microbiome interact directly with the gastrointestinal (GI) tract, but it also interacts with the immune system that is present in the GI tract and with the neurological system through various signaling systems. Gastrointestinal signaling is mediated in part by microbial metabolites and is involved in regulating the gut–brain axis in the host. (85) This section focuses on the bacterial microbiome by first discussing the techniques used to identify and study the gut microbiome. Then, we will discuss the human GI tract from an ecological viewpoint. The phyla commonly found in the gut microbiome will be defined and discussed. Finally, we will focus on probiotics and related compositions (prebiotics, postbiotics, synbiotics, and psychobiotics) and present recent examples of each class to illustrate the current state of the art.

Techniques Used to Study the Gut Microbiome

Until recently, about 400 bacterial species were identified in the human microbiome using conventional culturing techniques. (86) These techniques, by their nature, underestimate the actual number of species because, to culture a bacterium successfully, one needs to provide the correct nutrients, pH, and redox environment to enable growth. Conventional culture techniques favor fast-growing and nonfastidious species over those present in low concentration, which requires unusual culture conditions and/or complex nutritional requirements. (86) Most isolation methods use selective agents, such as bile salts, to enrich the numbers of a desired bacterial type over others in the sample. The choice of the proper selective agent then becomes important. Some gut bacteria depend on other microorganisms in the gut to provide the nutrients they require for growth “cross-feeding.” Devising culture media for these can be a hit-or-miss proposition. Some bacteria will only grow in niches that have a narrow range of pH and/or redox potential, which may be difficult to maintain in vitro. Although there have been methodological advances in anaerobic culturing techniques, they tend to be tedious, time-consuming, and require specialized equipment. (31) The successful culture of strict anaerobic bacteria requires training, experience, and careful planning. Additionally, some bacteria might be alive but unculturable. Intercellular adherence may reduce the number of organisms that can give rise to colonies. (87)
The development of culture-independent metagenomic approaches, such as 16S RNA gene sequencing and high-throughput sequencing, have been an enabling factor in the study of the human gut microbiome. This topic was reviewed in depth by Sankar et al. in 2015. (86) The 16S rRNA gene exhibits several advantages, including its distribution in all bacterial species, its absence in eukaryotes, its stability over time, and its size (∼1500 bp), that make it suitable for bioinformatic analyses. High-throughput sequencing methods have given unprecedented access to the analysis of the microbial diversity of complex microbiotas, particularly through metagenomic approaches. Two strategies used are high-throughput sequencing of pooled PCR-amplified 16S rRNA and shotgun sequencing of all DNA fragments present, which enables identification of the microorganisms present and their metabolic genes. With these molecular techniques, it is now estimated that the human GI microbiota comprises more than 2000 species using modern molecular methods. (86,88)
Recent impressive advances in next generation sequencing technologies, along with the progress and innovations of metagenomics, metabolomics, multiomics, bioinformatics, and artificial intelligence tools, have provided prospects to better characterize the microbial populations and their functions and help in better correlation prediction.

Gastrointestinal Tract

The gut microbiome varies according to the GI anatomy, which varies in terms of physiology, pH and O2 tension, flow rates (rapid from the mouth to the cecum, slower afterward), substrate availability, and host secretions. (89) Figure 6 presents a representation of the GI tract with some of the bacterial taxa present. The GI tract consists of the stomach, duodenum, jejunum, ilium, cecum, and colon, with each environment ascending in pH and growing progressively more anaerobic from stomach to colon. Each section of the GI tract presents a unique ecological niche that exerts selective pressure on the microbiome. It is an open system with nutrients entering the system intermittently and wastes also leaving intermittently. The microbiome is affected by many factors, including diet, medications (especially antibiotics), ethnicity, age, and general health. (82,83)

Figure 6

Figure 6. Gut-microbiota-participant bacteria.

The stomach is an extreme habitat because it is highly acidic (pH = ∼1.5). Once, it was considered sterile because of its acidity until the discovery of Helicobacter pylori in this hostile environment in 1982. The microbial population in the gastric environment is low and in the range of 101–103 cells/mL. (87) Investigations since this discovery have revealed that the gastric fluid is predominated by the members of Firmicutes, Bacteroidetes, and Actinobacteria. (87) The gastric mucosa was found to have a rich diversity with bacterial members belonging to Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, and Actinobacteria. In healthy human stomach, the genera Streptococcus, Prevotella, Veillonella, Rothia, and Haemophilus were found to be predominant; however, the composition of the gastric microbiota is dynamic and affected by such factors as diet, drugs, and diseases. (90)
The small intestine comprises the duodenum, jejunum, and ileum. The duodenum has a pH of 5–6.8. Bacterial numbers are 103–104 cells/mL where Firmicutes predominate. (91) The jejunum and ileum have a higher pH (6–8) with a 104–108 cell/mL density that comprises strict to facultative anaerobic Gram-positive and Gram-negative bacteria. The small intestine is lined with simple columnar epithelial tissue, which is covered by a mucus layer, and has a large surface area because of the villi and microvilli. When food enters the duodenum, the pH and bacterial load are low. These small intestinal mucosae are associated with members of phyla Bacteroidetes and Firmicutes. Food is blended with bile, bicarbonate, and digestive enzymes in the duodenum, and when the intestinal contents reach the large intestine, the food blend has been converted to a neutral to alkaline pH. The small intestine provides a more challenging environment for microbial colonizers given the short transit times (3–5 h) and the high bile concentrations. (83,91)
The large intestine consists of the cecum and colon and is characterized by slow flow rates and pH varying from 6 to 7.8. It harbors by far the largest microbial community. The large intestine is strictly anaerobic, and the cell density reaches 1012 cells/mL. The large intestine is home to the most complex bacterial diversity in the GI tract because of several factors, such as its larger volume, moderate or less acidic pH, low concentration of bile salts, and the longer retention time caused by slower peristalsis. Five major phyla─Firmicutes, Bacteroidetes, Actinobacteria, Verrucomicrobia, and Proteobacteria─covering a wide range of bacterial genera─Clostridium, Fusobacterium, Bacteroidetes, Actinomyces, and Propionibacterium─are associated with the large intestine. Other Gram-positive cocci─Micrococci, Peptococci, Peptostreptococci, and Ruminococci─have been also reported to play crucial roles in the large intestine. Food that has not been degraded in the upper GI tract reaches the large intestines and supports the microbiota with nutrients and energy. The carbohydrates present are fermented to carbon dioxide, hydrogen, methane, and short-chain fatty acids (SCFA) (primarily acetate, propionate, and butyrate). Most of the SCFA produced in the large intestine are absorbed by the host and provide an energy source. The amount of energy derived from SCFA accounts for 6–9% of the total energy requirement. (82,88,91,92)
The microbiome composition of the intestinal lumen, known as mucosal and epithelial spaces of the GI tract, is highly diverse and comprises Verrucomicrobia, Fusobacteria, Asteroplasma, Cyanobacteria, Actinobacteria, Lentisphaera, Spirochaetes, Bacteroidetes, Proteobacteria, Bacilli, Clostridia, and Mollicutes. The predominating genera are Escherichia, Klebsiella, Enterococcus, Bacteroides, Ruminococcus, Dorea, Clostridium, Coprococcus, Weisella, and Lactobacillus. Other genera found include Granulicatella, Streptococcus, and Veillonella. (83,91)

Types of Bacteria Found in the GI Tract

The four dominant phyla residents in the human gut are Firmicutes (which contains lactobacilli), Bacteroidetes, Actinobacteria (which contains Bifidobacteria), and Proteobacteria. Other phyla found in lower numbers are the Fusobacteria and Verrucobacteria. Figure 7 presents the significant phyla, families, and genera of gut bacteria in terms of the number of records that cite them in the CAS Content Collection. This presentation reflects the relative level of research interest in each of these taxonomic groups. Most bacteria belong to the genera Bacteroides, Clostridium, Fusobacterium, Eubacterium, Ruminococcus, Peptococcus, Peptostreptococcus, and Bifidobacterium. Other genera, such as Escherichia and Lactobacillus, are present to a much lesser extent. Twenty-three species from the genus Bacteroides, alone, constitute about 30% of all bacteria in the human gut. (93)

Figure 7

Figure 7. Representation (as number of records) of the gut bacteria phyla and species in the CAS Content Collection.

Bacteroidetes

Bacteroidetes are Gram-negative, nonspore-forming, anaerobic or aerobic, rod-shaped bacteria. Bacteroides fragilis, found in the human microbiome, is the type of species for this phylum. The majority of the Bacteroidetes species fall into three genera: Prevotella (bile-sensitive, moderately saccharolytic, with pigmented and nonpigmented species), Porphyromonas (bile-sensitive, pigmented, asaccharolytic species), and Bacteroides (bile-resistant, nonpigmented, saccharolytic species). Other genera in the phyla are Alistipes, Anaerorhabdus, Dichelobacter, Fibrobacter, Megamonas, Mitsuokella, Rikenella, Sebaldella, Tannerella, and Tissierella. (94) Some members of the Bacteroides genus, although belonging to the normal gastrointestinal microbiota, can cause opportunistic infections if the integrity of the intestinal mucosal barrier is broken. These infections are usually polymicrobial, but B. fragilis and B. thetaiotaomicron are the most frequent species isolated. Some members of the genera Porphyromonas, Prevotella, and Tannerella are well-known pathogens of the oral cavity, where they can notably cause periodontal disease and dental caries. (95) The ability of some members of the Bacteroidetes to degrade polysaccharides explains why they thrive in the GI tract. (96)

Firmicutes

The Firmicutes phylum comprises Gram-positive bacteria with low G + C DNA content and is composed of more than 200 different genera, such as Lactobacillus, Bacillus, Clostridium, Enterococcus, and Ruminicoccus. (82) They can be found in a variety of places, including soil, water, skin, and the GI tract. The phylum includes aerobes, anaerobes, spore-forming, saprophytic, and pathogenic bacteria. Notable among the latter are Clostridium difficile and Listeria monocytogenes. Firmicutes, such as Clostridium botulinum, Clostridium tetani, Clostridium perfringens, and Staphylococcus aureus, can produce proteinaceous toxins. Other important genera are Listeria, Paenibacillus, Staphylococcus, Streptococcus, Pediococcus, and Leuconostoc. (97) Some members of Firmicutes are involved in bile acid metabolism in the gut. Accumulating evidence suggests that bile acids play pivotal roles in gut inflammation and the development of intestinal bowel disease (IBD). Patients with IBD exhibit decreased microbial diversity and abnormal microbial composition marked by the depletion of phylum Firmicutes. (98)

Actinobacteria

The Actinobacteria are Gram-positive bacteria with high G + C DNA content and constitute one of the largest bacterial phyla. They are ubiquitously distributed in both aquatic and terrestrial ecosystems. Many Actinobacteria have a mycelial lifestyle and undergo complex morphological differentiation. They also have an extensive secondary metabolism and produce about two-thirds of all naturally derived antibiotics in current clinical use, as well as many anticancer, anthelmintic, and antifungal compounds. The phylum includes pathogens (species of Corynebacterium, Mycobacterium, Nocardia, and Propionibacterium), soil inhabitants (Micromonospora and Streptomyces species), plant commensals (Frankia spp.), and GI commensals (Bifidobacterium spp.) (99) The Bifidobacteria are among the first microbial colonizers of the intestines of newborns and play key roles in the development of their physiology, including maturation of the immune system and use of dietary components. Some Bifidobacterium strains are considered probiotic microorganisms because of their beneficial effects, and they have been included as bioactive ingredients in functional foods, mainly dairy products, as well as in food supplements and pharma products, alone or together with other microbes or microbial substrates. (100)

Proteobacteria

The name Proteobacteria was first proposed by Stackebrandt et al. in 1988. (101) The name was derived from Proteus the ancient Greek god of the sea capable of assuming different shapes, which reflected the high heterogeneity displayed by the bacteria belonging to this phylum. A common trait of Proteobacteria is Gram-negative staining, which indicates the presence of lipopolysaccharide in the outer membrane. On the basis of phylogenetic analysis of the 16S rRNA gene, the Proteobacteria phylum is divided into six classes (previously regarded as subclasses of the phylum): Alphaproteobacteria, Betaproteobacteria, Gammaproteobacteria, Deltaproteobacteria, Epsilonproteobacteria, and Zetaproteobacteria. Considering that the classes division is based on molecular relatedness, it is not surprising that no specific morphological or physiological trait characterizes members within each class. (102) Notable genera in the Proteobacteria are Escherichia, Salmonella, Shigella, Desulfovibrio, and Helicobacter. Included in the phyla is the Enterobacteriaceae family, which contains several enteropathogenic bacteria, including Shigella flexneri, Salmonella typhi, and Escherichia coli. Other enteric pathogens in this phylum are Vibrio cholerae and Helicobacter pylori.

Verrucomicrobia

The phylum Verrucomicrobia, like the Proteobacteria, is defined by as a distinct phylogenetic lineage, as determined by 16S rRNA gene sequences. The phylum has been recognized as separate since 1995 but currently counts only a few cultivated microorganisms as members. Verrucomicrobia is a divergent phylum that includes members of the microbial communities of soil and fresh and marine waters. Some extremely acidophilic members from hot springs have been found to oxidize methane. (103,104) Akkermansia muciniphila is a mucus-degrading member of the Verrucomicrobia found in the human GI tract. A. muciniphila represents from 1 to 4% of the bacterial population in the colon. (105) A. muciniphila prefers to colonize in the intestinal mucus layer and specifically degrades mucins to produce short-chain fatty acids, thereby providing energy for the host and promoting colonization of the bacterium itself. The degradation of mucins prompts the host to compensate with the production of more mucins, thereby maintaining the dynamics of these proteins. (106)

Fusobacteria

The phylum Fusobacteria is made up of Gram-negative, nonmotile, facultative aerobic to obligately anaerobic, fermentative, rod-shaped bacteria, which have generally fusiform (spindle-shaped) morphology. Fusobacteria have been known for more than 100 years, but recently, phylogenetic studies have shown that they should be grouped into a distinct phylum. The bacteria from this phylum are commonly associated with the mucous membrane of humans and animals. They are also commonly present in the human and animal GI tract, particularly in the jejunum, the ileum, and the colon. (83,107)

Gut Microbiome–Disease Correlations

The human microbiome has been recognized as an essential factor for human health. (108−110) Specifically, gut microbes contribute directly and/or indirectly to important physiological activities, including immunomodulation and the regulation of various neurotransmitters, hormones, and metabolites. Dysbiosis is a state characterized by distinct alterations in the microbiome that result in an imbalance in the microbiota, modifications in their functional composition and metabolic performance, or a change in their allocation. The impact of the microbiome on human physiology and pathology is so extensive that the microbiome has been considered as an essential organ of the human body. (111−113) A search in the CAS Content Collection identified a large collection of studies reporting correlations between gut microbiota and a wide range of diseases, including mental, metabolic, and digestive system disorders; cardiovascular and neurodegenerative diseases; various cancers; and immune and autoimmune diseases (Figure 8). Trends in the number of publications related to various diseases in the recent years (2016–2021) are depicted in Figure 9. The number of documents related to dysbiosis, in general, exhibits the greatest growth rate, thereby characterizing the dominant fundamental approach of the recent studies in the field.

Figure 8

Figure 8. Distribution of the publications in the CAS Content Collection related to gut microbiome-associated diseases.

Figure 9

Figure 9. Trends in the number of publications concerning gut microbiome-related diseases during the years 2016–2021. Percentages are calculated with yearly publication numbers for each disease, normalized by the total number of publications for the same disease in the same time period.

Digestive System Diseases and Disorders

Alterations to the gut microbiota composition have been associated with various digestive system disorders and diseases, specifically IBS; IBD, including Crohn’s disease and ulcerative colitis; diarrhea, and constipation (Figure 8). (114−116)
Irritable bowel syndrome is one of the most prevalent functional gastrointestinal disorders and is considered as the prototype of disorders of the gut–brain interaction. While alterations in gut–brain interactions have clearly been established in IBS, a causative role of the microbiome remains to be determined. Dysbiosis is one of the hallmarks in the miscommunication between gut and brain and could lead to IBS symptoms. The severity of IBS symptoms has been shown to be correlated with dysbiosis. (117,118) In IBS cases, the reduction of microbiome diversity, gut barrier deficiency, gut–brain signaling disorders, and immune disorders are significantly related to the abnormal function of the GI tract. (119) Modifications in the composition of the normal microbiota and perturbed colonic fermentation in IBS patients are supposed to play a role in the development of IBS, with a considerable, nearly 2-fold, increase in the ratio of Firmicutes to Bacteroidetes. (120) Recent studies have reported well-defined distinction between gut microbiota composition in patients with IBS compared with healthy controls. IBS was typified by enhanced quantities of Firmicutes and specifically in Ruminococcus, Clostridium, and Dorea, along with a distinct decrease of beneficial microbes, such as Bifidobacterium and Faecalibacterium spp. (121) Moreover, a decrease in probiotic species and an increase in pathogenic species have been reported in patients with IBS, including Proteobacteria, Enterobacteriaceae, Lactobacillaceae, and Bacteroides (Bacteroidetes). (122) Fecal transplantation from super donors and microbiome modulation either by pro- or prebiotics have shown beneficial effect in reducing IBS symptoms and improving patients’ quality of life. (119,123,124) To date, the guidelines on the treatment of IBS with probiotics remain controversial. The British Society of Gastroenterology guidelines (125) on the management of IBS, which were updated in 2021, reported that probiotics may be an effective treatment for improving global symptoms and abdominal pain in patients with IBS, which was consistent with the recommendations of the Canadian Association of Gastroenterology (126) and the Japanese Society of Gastroenterology. (127) In contrast, the guidelines from the American College of Gastroenterology (128) suggest against the use of probiotics for the treatment of global IBS symptoms. (129)
Like IBS, inflammatory bowel disease-related dysbiosis is associated with a general decrease in richness, diversity, and stability of the microbiota. (130) This decline in diversity is concomitant with a weakened immune response and setbacks with the cellular barrier functions that normally block bacterial entry from the gut lumen into gut tissue. These malfunctions trigger complications with antibacterial defense and consequent growth of pathogenic bacteria. (131) IBD-related dysbiosis is specifically associated with a comprehensive decrease in the quantity and diversity of Firmicutes and an increase in Proteobacteria. (132) The decrease in the numbers of Firmicutes is noteworthy since they produce essential short-chain fatty acids, such as acetic and butyric acids, that are known to exhibit anti-inflammatory properties. (133) A common feature of the microbial dysbiosis among IBD patients, especially in Crohn’s disease, is the decreased abundance of Firmicutes bacteria belonging to two families that are important functional members of the human gut microbiota─Ruminococcaceae and Lachnospiraceae─to which most butyrate-producing bacteria in the human gut belong. (134,135) Thus, depletion of these bacterial families in IBD is supposedly correlated to the detected disturbances, such as a lower butyrate-producing capacity of the IBD microbiota. (136) Butyrate has a significant potential in IBD therapy because it serves as the colonocytes key energy source, enhances the epithelial barrier integrity, and inhibits inflammation. A probiotics treatment, including consumption of butyrate-producing bacteria to increase in situ butyrate production, may restore gut homeostasis. (137−139) A recent study reported that an orally delivered cocktail of bacteriophages targeting an IBD-associated strain of the bacterium Klebsiella pneumoniae alleviated intestinal inflammation. (131)
A growing body of evidence shows that imbalance of the gut microbiota increases susceptibility to various pathogens and causes numerous diseases, including diarrhea. (140) At present, the pathogens causing diarrhea are believed to be Escherichia coli, Shigella, Salmonella, Campylobacter, Clostridium difficile, and Aeromonas. (141,142) It has been found that microbial intervention can regulate the composition of the intestinal flora to prevent and improve the occurrence of diarrhea. (143) Probiotics containing nonpathogenic live bacteria preparations, such as Lactobacillus, Yeast, Bifidobacterium, Enterococcus, and Bacillus, have been demonstrated to treat pathogens-caused diarrhea by preserving or amending the balance of gut microbiota. The mechanisms of the beneficial effect are supposedly related to the inhibitory effect on the colonization of pathogenic bacteria by competing for nutrients and producing antibacterial compounds. (144)
Accumulating evidence suggests an association between functional constipation and abnormal gut microbiota, with the relationship between gut microbiota and gut transit being likely bidirectional. (145) By controlling colonic motility, water content, secretion, and absorption, gut microbiota may promote the development of functional constipation through microbial metabolites, including bile acids, SCFAs, 5-hydroxytryptamine, and methane. Currently, there is no consensus on the gut microbial composition typical for functional constipation patients and the alteration trends of the various microbial classes compared with healthy controls. (146−150) However, recent studies showed that changes in the mucosal and fecal microorganisms are linked to functional idiopathic constipation. Taxonomic profiling of intestinal microbiota in constipated adults showed a higher abundance of Bacteroides and other pathogenic microorganisms than in healthy volunteers. (151) The increased richness and diversity of the gut microbiomes result in slow colonic transit. In addition, intestinal microbiota in constipated adults have genes involved in pathways that lead to methane, hydrogen, and glycerol production, which can explain the symptoms seen in patients with constipation. (151−153) Microbial interventions including probiotics, prebiotics, and synbiotics, which bring about compositional and functional changes of the gut microbiota, have frequently shown beneficial effects on functional constipation that are in favor of the concept of the significant role of gut microbiota in functional constipation. (145) This concept is supported also by the reports that many risk factors of functional constipation, including age, diet, obesity, and stress, have a considerable effect on the gut microbiota. (154,155)

Mental and Neurodegenerative Disorders

Gut microbiota have been reported to affect neurological functions along the so-called gut–brain axis (GBA). (156) Gut microbiota communicates with the brain through three major routes: the neural route (vagus nerve, enteric nervous system), the immune route (cytokines), and the endocrine route [hypothalamus–pituitary–adrenal (HPA) axis, gut hormones]. Disturbances in any of these routes can result in mental disorders. Dysbiosis in common intestine microbial species of the phylum Firmicutes and Actinobacteria and the genera Bacteroides and Bifidobacterium are supposedly responsible for mental health disorders. (157) Gut microbiota moderate the GBA via various ways, such as preserving gut permeability by controlling the integrity of tight junctions in gut epithelium and producing a large selection of metabolites that include neurotransmitters, SCFAs, and amino acids.
A plethora of research reports have indicated the significance of microbiota in the development of neurodegenerative diseases via a variety of microbial metabolites that transmit from the gut to the brain across the GBA. (158,159) Changes in the levels of gut microbial metabolites have been reported to be associated with neurological conditions like Parkinson’s disease, (160) anorexia nervosa, (161) Alzheimer’s disease, (162) autism spectrum disorders, (163) and chronic stress and depression. (164) It is not clear by now, however, whether these disruptions in mental health are the cause or a result of the changes in gut microbiota. Gut dysbiosis has been associated with increased gut permeability and inflammation and it may also cause enhanced levels of circulating gut microbiota metabolites, such as the neurotoxin β-N-methylamino-l-alanine and microbial amyloids. (165,166) β-N-Methylamino-l-alanine is one of the gut cyanobacteria-produced neurotoxins causing neurodegeneration, cognitive impairment, and the accumulation of neurofibrillary tangles. (167,168) The hypothesis that Parkinson’s disease starts in the gut and spreads to the brain (169) is gaining increasing support, thereby showing that the disease is associated with widespread dysbiosis. (170)
Alterations in the microbiota composition in patients with Alzheimer’s disease compared with matched healthy controls included a reduction in richness and diversity of gut microbiota with decreased Firmicutes and Bifidobacterium and increased Bacteroidetes. (171) Changes in Actinobacteria, Ruminococcus, Lachnospiraceae, and Selenomonadales (1686) have also been reported. (172) Cognitively impaired patients exhibited alterations in Bacteroidetes, Firmicutes, Proteobacteria, and Verrucomicrobia compared with age-matched cognitively intact individuals. (173)
Therapeutic interventions including the administration of pre- and probiotics (psychobiotics) to manage mental disorders and/or their symptoms have been undertaken. (174,175) These inventions have included probiotic combinations of lactobacilli and Bifidobacteria, which has resulted in a significant drop in psychological distress, (176) enhanced cognition and communication among patients with Alzheimer’s disease (177) and autism spectrum disorders, (178) and recovering symptoms among patients with Parkinson’s disease. (179) On the basis of the promising results of psychobiotics on controlling or modulating the GBA, additional clinical trials are currently being undertaken to identify bacterial strains as promising candidates for the treatment of mental disorders.
Humans are adapted to a circadian rhythm of 24 h associated with the light/dark cycle on earth. The central circadian clock is located in the hypothalamus, which synchronizes information on environmental light and dark signals to peripheral tissues to keep the body functioning in the same rhythm. (180) A disruption of circadian rhythms is associated with various diseases, including neurodegenerative diseases, sleep, and psychiatric disorders. (181,182) Recent studies have reported that gut microbiota are able to control or be controlled by the circadian clock. The mechanisms of such a relationship requires small molecule gut microbiota metabolites, such as bile acids and SCFAs, to act as intermediaries. (180) Thus, the levels of butyrate and propionate show obvious daily oscillations. Moreover, these oscillations are lost under high-fat diets. (183) The impacts of gut microbiota metabolites on circadian rhythm are extensive and are connected to other functions of gut microbiota metabolites, such as energy metabolism and immunity. These interconnections between different physiological functions via the link of gut microbiota metabolites are essential for understanding the functions of gut microbiota metabolites and the general role of gut microbes in human health and disease.

Metabolic Disorders

Systemic metabolic diseases that are believed to be strongly affected by gut microbiota status include obesity and diabetes. (184) Gut microbial composition is strongly affected by dietary routines. As a result of a high-fat diet, the intestinal microbiome is modified with rising amounts of Firmicutes and Proteobacteria and reduced levels of Bacteroidetes. The Firmicutes/Bacteroides ratio has been correlated to body weight, which means it is larger for obese people. (185) Clostridium difficile infections can also trigger obesity. Generally, obesity is affected by the inflammatory status induced by gut bacteria or their metabolites, which regulate the GBA. (108,185)
Diabetes is another metabolic disease that is strongly associated with the gut microbiome. Studies have reported an increased quantity of Villanella, Clostridium, and Bacteroides and a decreased quantity of Lactobacillus, Eubacterium rectale, Blautia coccoides, and Bifidobacterium in children with type 1 diabetes. Besides, negative correlation has been reported between plasma glucose level and Bifidobacterium, Lactobacillus spp., and Firmicutes and Bacteroidetes spp., while there has been positive correlation between Clostridium and plasma glucose level. The ratios of Bacteroidetes to Firmicutes were reported to exhibit a positive connection with plasma glucose levels. The Lactobacillus genus was also in lower quantity in type 2 diabetes patients, and Bifidobacterium was in higher quantity compared with control groups. (108,186) Risks for the development of type 2 diabetes have been correlated to the composition of gut microbiota, as well. The alterations in the gut microbiota of individuals with type 2 diabetes have been small compared with the control group, yet a consistent decline in the metabolically beneficial butyrate-producing bacteria was reported. (186) Overall, type 2 diabetes was associated with a reduced quantity of SCFAs-producing bacteria, in particular butyric acid, which has been related to insulin sensitivity. (187,188) The relation between SCFAs and insulin sensitivity stems from the capacity of SCFAs to stimulate the secretion of GLP-1 by intestinal L-cells via G protein receptors, which has a significant impact on insulin release. (189)
Close relations between the metabolic and immune systems are now largely supported, and intestinal microbiota is being progressively identified as an important factor connecting genes, environment, and the immune system. (190)

COVID-19

Recently, correlation has been reported between gut microbiota composition and levels of cytokines and inflammatory markers in patients with COVID-19. (191,192) It is suggested that the gut microbiome is involved in the magnitude of COVID-19 symptoms’ severity via modulation of the host’s immune responses. Moreover, gut microbiota dysbiosis could contribute to persistent symptoms even after disease resolution, thereby emphasizing a need to understand how gut microorganisms are involved in inflammation and COVID-19.A recent study demonstrated that SARS-CoV-2 infection indeed disrupts the gut microbiome. (193) This boosts secondary bacterial infections both by facilitating pathogenic bacteria to colonize the gut and by modifying gut lining to allow these bacteria to spread from the gut to the bloodstream of COVID-19 patients. These results confirm the direct role of gut microbiome dysbiosis in facilitating grave secondary infections upon COVID-19 malady. (193)
The alterations to the gut microbiota composition related to digestive system diseases, mental health, and metabolic disorders are summarized in Table 1.
Table 1. Gut Dysbiosis in Digestive System Diseases, Mental Health, and Metabolic Disorders
diseases ↓ decreasing bacteria ↑ increasing bacteria
digestive system diseases
irritable bowel syndrome (194−199) Bifidobacterium Ruminococcus
Faecalibacterium prausnitzii Dorea
Bacteroides Enterobacteriaceae
Lactobacillaceae
Bacteroides
Firmicutes/Bacteroidetes ratio
IBD: Crohn’s disease (200,201) Bacteroides  
Faecalibacterium prausnitzii
Bifidobacterium adolescentis
IBD: ulcerative colitis (139,200) Bifidobacteria  
Roseburia hominis
Faecalibacterium prausnitzii
Lachnospiraceae
Ruminococcaceae
mental health disorders
anxiety disorder (202,203) Bacteriodetes Bacteroidaceae
Ruminococcus gnavus Enterobacteriaceae
Fusobacterium Burkholderiaceae
post-traumatic stress disorder (204) Actinobacteria  
Lentisphaerae
Verrucomicrobia
depression (205−207) Prevotella Eggerthella
Dialister Holdemania
Turicibacter
Paraprevotella
dementia (172) Actinobacteria Escherichia
Bacteroides Blautia
Bifidobacterium
Streptococcus
Lactobacillus
Dorea
metabolic disorders
diabetes type 1 (208,209) Lactobacillus Clostridium
Bifidobacterium Bacteroides
Blautia coccoides Veillonella
Eubacterium rectale Actinobacteria
Prevotella Proteobacteria
Akkermansia Lactococcus
Firmicutes  
diabetes type 2 (199,209,210) Firmicutes Betaproteobacteria
Clostridia  
Lactobacillus Bacteroidetes/Firmicutes ratio
Akkermansia muciniphilia
Roseburia
obesity (199,211) Bacteroidetes Enterobacteria
Methanobrevibacter smithii Ruminococcus gnavus
Ruminococcus flavefaciens Actinobacteria
Bifidobacterium Prevotellaceae

Gut Bacteria–Disease Correlations

In an effort to get better insight into the gut microbiota impact on well-being, we explored the correlations between the major classes of gut bacteria and certain mental and gastrointestinal disorders, as reflected in the number of records in the CAS Content Collection (Figure 10).

Figure 10

Figure 10. Correlation between major classes of gut bacteria with mental and gastrointestinal disorders, as reflected in the number of associated records in the CAS Content Collection.

As seen from Figure 10, Bacteroidetes are the most studied class of gut bacteria with relation to gastrointestinal and mental health, specifically with relation to stress, depression, and anxiety. Bacteroidetes are known to have a very broad metabolic potential and are regarded as one of the most stable parts of gastrointestinal microflora that exhibits remarkable nutritional flexibility and an ability to respond to stress. (212) However, the exact mechanisms underlying any possible relationship between Bacteroidetes and mental and gastrointestinal health remains unclear, and further research is needed to fully understand this complex relationship.
Firmicutes are the second extensively studied class of gut bacteria with relation to gastrointestinal and mental health, especially in relation to stress and depression (Figure 10). Many members of the Firmicutes phylum, such as Lactobacillus, are probiotic. The relationship between Firmicutes and gastrointestinal disorders may be mediated by a number of different factors, such as the production of SCFAs by Firmicutes, which are an important energy source for the gut epithelium and have anti-inflammatory effects. In addition, some Firmicutes bacteria are involved in the fermentation of complex carbohydrates and the production of beneficial metabolites, such as butyrate, which has been shown to have protective effects against colorectal cancer. (213) Like with Bacteroidetes, the exact relationship between Firmicutes and mental and gastrointestinal disorders is still being studied and is not fully understood.
The interest in Fusobacteria with respect to the gastrointestinal and mental health is mainly related to diarrhea and constipation (Figure 10). Recent evidence is emerging that this bacterium may be related to human colon cancer. (214)
It is noteworthy that recent meta-analysis has reinforced the genetic correlations between Alzheimer’s disease and the gut microbiome genera. (215) For example, genus Actinobacterium Collinsella was confirmed to be associated with Alzheimer’s disease, as well as rheumatoid arthritis, atherosclerosis, and type 2 diabetes. (215) It is also worth reiterating that the gut microbiota is a complex and diverse community of microorganisms, and changes in any one particular phylum are unlikely to fully explain the development of any particular disorder or condition. Rather, the gut microbiota as a whole are likely to play a role in shaping our physical and mental health, and further research is needed to fully understand the complex relationships between the gut microbiota and overall well-being.

Therapeutic Strategies for the Treatment of Mental and Gastrointestinal Disorders

Imbalances within the gut microbiome–brain axis have been linked to a range of mental and gastrointestinal disorders. Assorted therapeutic interventions aimed at modulating the gut microbiome or the gut–brain axis may be effective in improving outcomes for these conditions.
Dietary Interventions
A high-fiber diet can increase the production of SCFAs, which can help to maintain gut barrier function and reduce inflammation. SCFAs can also promote the growth of beneficial gut bacteria, which can outcompete pathogenic bacteria and reduce inflammation. (216)
A diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) can reduce symptoms in individuals with irritable bowel syndrome by reducing the fermentation of certain carbohydrates in the gut, which can cause symptoms such as bloating and abdominal pain. However, the low-FODMAP diet can also reduce the diversity of gut bacteria and may have negative long-term effects on gut health. (217,218)
Probiotics and Prebiotics
Probiotics can improve gut barrier function by enhancing the production of mucus and tight junction proteins, which can prevent the entry of harmful molecules and pathogens into the bloodstream. They can also reduce the production of proinflammatory cytokines and modulate the activity of immune cells in the gut, thereby promoting an anti-inflammatory response. (219,220)
Prebiotics can increase the production of metabolites, such as SCFAs, which are important energy sources for gut cells and can help to maintain gut barrier integrity. SCFAs can also activate G protein-coupled receptors on immune cells, thereby leading to the production of anti-inflammatory cytokines and the suppression of proinflammatory cytokines. (221,222)
Antibiotics
Antibiotics can kill harmful bacteria in the gut, which reduces inflammation and restores gut barrier function. However, antibiotics can also have negative effects on the gut microbiome, such as by reducing the diversity of gut bacteria and promoting the growth of antibiotic-resistant bacteria. Antibiotics should be used judiciously and only when necessary. (223,224)
Fecal Microbiota Transplantation (FMT)
FMT can restore the composition and function of the gut microbiome, which can reduce inflammation and improve gut–brain communication. FMT has been shown to be effective in treating recurrent Clostridium difficile infection and is being investigated for other conditions, such as IBD and autism. (225−227)
Psychotherapeutic Interventions
Psychotherapeutic interventions can reduce stress and improve gut–brain communication. Stress can disrupt gut microbiome composition and function, which leads to inflammation and the development of diseases related to the gut microbiome–brain axis. The reduction of stress can be an effective way to improve gut health and reduce symptoms in these conditions. (228−230)
Pharmacological Interventions
Drugs that target the gut–brain axis can modulate gut microbiome composition and function and reduce inflammation. For example, certain drugs that target the serotonin system, such as selective serotonin reuptake inhibitors (SSRIs), can modulate gut–brain communication and have been shown to be effective in treating conditions, such as depression and anxiety. (231,232)
Figure 11 demonstrates the annual growth in the number of documents in the CAS Content Collection related to various therapeutic interventions applied for the treatment of mental and gastrointestinal disorders. Fecal transplant as a new therapeutic strategy exhibits the fastest growth rate recently.

Figure 11

Figure 11. Trends in the therapeutic strategies applied for the treatment of mental and gastrointestinal disorders, as presented in the documents related to gut microbiome research during the years 2017–2021. Percentages are calculated with yearly publication numbers for each type of therapeutic intervention normalized by the total number of publications for the same intervention in the same time period.

The various strategies to treating diseases related to the gut microbiome–brain axis can work through multiple mechanisms, including modulating gut microbiome composition and function, reducing inflammation, and improving gut–brain communication. (233) However, the underlying mechanisms are complex and require further research to fully understand.

Gut Microbiota Metabolites

ARTICLE SECTIONS
Jump To

It is estimated that the human gut microbiome contains more than 22 million microbial genes, (234) which exceeds the ∼22,000 genes present in the entire human genome. (235) These genes enable the gut microbiota in the host to synthesize a myriad of enzymes with versatile capabilities to ferment and degrade a variety of compounds that humans do not have the genetic machinery to metabolize. As a result, the gut microbiota can generate a battery of metabolites with a wide spectrum of bioactivities. The gut microbiota-derived metabolites can be broadly divided into three types according to their origination: (1) metabolites that are produced by gut microbiota directly from diets; (2) metabolites that are generated by the host and modified by gut microbiota; (3) metabolites that are produced de novo. (236) A selection of important gut microbiota metabolites related to gut–brain communication is shown in Table 2.
Table 2. Exemplary Gut Microbiota Metabolites, as Represented in the CAS Content Collection

Gut Microbiota Metabolites with Impact on Brain Function

The gut microbiota provides essential signaling metabolites that are vital for the host’s physiology. While in healthy individuals, gut microbiota metabolites are effective in maintaining the important functions of hosts, perturbations in the production of these metabolites can initiate various diseases, such as digestive system diseases, neurodegenerative and metabolic disorders, and cancer. (236)
Research has shown that gut microbiota is crucial for normal brain development and function. (237,238) For example, administration of 4-ethylphenylsulfate (4-EPS), a tyrosine derivative, to mice at 3–6 weeks postnatal induced anxiety-like behavior. The biosynthetic pathway analysis and mechanisms behind the detrimental effects of 4-EPS showed that 4-EPS interfered with oligodendrocyte maturation, myelination, and brain activity patterns. p-Cresol, another tyrosine derivative and a metabolite, has also been directly associated with neurodevelopmental disorders. (239,240) Further, certain bacteria-related metabolites, such as trimethylamine-N-oxide (TMAO), 5-aminovaleric acid (5-AVA), 5-AVA betaine (5-AVAB), imidazolepropionic acid, and hippuric acid have been reported to promote early-life axonogenesis both in vitro and in vivo. (241) Moreover, the neurogenic properties of microbe metabolites may not be limited to early life given that indole, a tryptophan metabolite, has been reported to increase neurogenesis in the hippocampus of adult mice. (242) Pilot studies using fecal microbiota transplantation in children enabled the assessment of whether early life interventions affecting the gut microbiota composition exerted long-term neurodevelopment effects. (243,244) Thus, maternal fecal microbiota transplantation in cesarean-born infants was found to rapidly restore normal gut microbial development. (244)
Gut microbiota and its metabolites can affect the host metabolism of neuroactive compounds. (245) The foremost examples of gut bacteria-derived neurotransmitters are aromatic amino acid derivatives dopamine and norepinephrine and glutamate derivative γ-aminobutyric acid. (246,247) Microbiota have been found to extensively contribute to the levels of dopamine and norepinephrine via the activity of β-glucuronidase. (248) Kynurenic acid, a tryptophan metabolite, functions as a glutamate modulator to reduce glutamate levels in the glutamatergic signaling in the hippocampus. Thus, enhanced cognitive abilities and memory in model animals have been achieved as a result of enhancing glutamate levels via limiting hippocampal kynurenic supply. (249,250) Over 90% of serotonin in the body is known to be produced in the gut in a process in which gut microbes play an important regulatory role. (251,252) Tyramine, deoxycholic acid, and 4-aminobenzoic acid have been reported to stimulate serotonin synthesis. (252) Furthermore, microbiota-related metabolites, such as norepinephrine, indole, indole-3-aldehyde, isovaleric acid, butyric acid, and isobutyric acid stimulate serotonin release from enterochromaffin cells. (253,254) Another gut microbiota metabolite with a likely connection to γ-aminobutyric acid (GABA) expression in brain is lactate. (255,256) It has been shown to affect neural plasticity and has a beneficial effect on learning and memory in model animals. (257) SCFAs, particularly butyric acid, may also have additional regulatory effects on the signal transduction to the brain via the vagal nerve and by inducing the biosynthesis of neurotransmitters in the CNS. (258) Administration of SCFAs, such as butyric, acetic, and propionic acid, has been reported to improve stress response, anxiety, and depression. (259) The presence of pipecolic acid in the CNS can be partially derived from the gut microbiota and has been also associated with GABA signaling and release. (245,260,261)
Gut microbiota has also proven vital for normal BBB function, especially during pre- and postnatal periods. (262) In a mice model of traumatic brain injury typified by acute BBB disruption, sodium butyrate administration exhibited an alleviating effect on BBB integrity. (263) Propionic acid, another SCFA gut microbiome metabolite, has also been shown to promote BBB integrity by mitigating oxidative and proinflammatory pathways. (264) It has been suggested that the effects of SCFAs on BBB integrity may rather be brought about by peripheral signaling instead of direct uptake to the brain, as implied by animal models. (258) Secondary bile acids, such as deoxycholic acid and ursodeoxycholic acid, may also modulate BBB integrity. (265,266) Trimethylamine, a metabolite of dietary choline, betaine, and l-carnitine, has been reported to exert detrimental impact on the BBB integrity. It is noteworthy that physiologically appropriate doses of the oxidized form of trimethylamine, TMAO, improved the BBB integrity. (267)
A large portion of the brain’s energy production is consumed by the neurons, the major component of the nervous tissue, in order to maintain the excitability of the synapses. (268) Lactate, a major gut microbiota metabolite, is known to augment neural activity as a primary energy source. (269) Modulation of brain energy metabolism in the hippocampus along the GBA is suggested to be responsible for improvement in the cognitive function after intermittent fasting in model animals. (270) It has been found that fasting considerably increased plasma levels of indolepropionic acid and tauroursodeoxycholic acid and fecal levels of SCFAs. Administrations of indolepropionic acid or tauroursodeoxycholic acid or a SCFAs mixture including acetic, propionic, and butyric acid have been able to reproduce the effects of fasting in cognition, hippocampal mitochondrial biogenesis, and energy metabolism-related gene expression. Findings connecting microbial metabolites to brain bioenergetics are preliminary but show that certain compounds are incorporated in the neuronal energy metabolism.
Gut microbiota metabolites reducing oxidative stress or neurotoxic proteins aggregation are functioning as neuroprotective agents. Metabolites that reduce inflammation or promote neurodevelopment or neurotransmission can also be considered as neuroprotectants. For example, ferulic acid is known to be metabolized by gut microbes. (271) It exerts neuroprotective effects by reducing neuronal cell death and recovers memory deficits in a cerebral ischemia and reperfusion injury model. (272) It has also ameliorated depressionlike behavior and oxidative stress. (273) Dihydroferulic acid, a microbiota metabolite, has also been shown to exhibit neuroprotective antioxidative properties. (274)
The gut microbiome metabolites and their mechanism of action in mental health and brain development are depicted in Figure 12, and their function and associated diseases are summarized in Table 3.

Figure 12

Figure 12. Exemplary gut microbiome metabolites and their mechanism of action in gut–brain communications.

Table 3. Gut Microbiota Metabolites, Their Function, and Associated Diseases
metabolite class/references specific functions associated diseases
short-chain fatty acids (258,285−290) – gut microbiota composition regulation – diabetes
– gut barrier integrity support – obesity
– energy homeostasis support – nonalcoholic fatty liver disease
– gut hormone production – ulcerative colitis
– circadian rhythm regulation – Crohn’s disease
– proinflammatory cytokines inhibition – colorectal cancer
– immunomodulation – autism spectrum disorder
– water, sodium, calcium, magnesium absorption – Parkinson’s disease
– regulation of intestinal pH value – diarrhea
– IBS
– constipation
– functional dyspepsia (FD)
bile acids (BAs) (291−295) – lipid and vitamin absorption regulation – obesity
– gut microbiota composition regulation – nonalcoholic steatohepatitis
– gut hormones production – ulcerative colitis
– intestinal immunity – cancer
– intestinal electrolyte and fluid balance – multiple sclerosis
– gut motility – Alzheimer’s disease
– gut barrier integrity – Parkinson’s disease
– lipid homeostasis – traumatic brain injury
– glucose homeostasis – stroke
– amino acid homeostasis – amyotrophic lateral sclerosis
– circadian rhythm – IBS
– neurotransmission  
tryptophan and indole derivatives (296−300) – gut microbial spore formation – ulcerative colitis
– drug resistance – Crohn’s disease
– biofilm formation – obesity
– intestinal barrier function regulation – stroke
– gut hormone secretion – mucosal candidiasis
– gut motility – autism spectrum disorder
– immunomodulation – Alzheimer’s disease
– Parkinson’s disease
– migraine
– schizophrenia
– IBS
choline metabolites (301−303) – bile acid synthesis inhibition – nonalcoholic fatty liver disease
– inflammation promotion – obesity
– thrombosis – diabetes
– myocardial hypertrophy and fibrosis – hypertension
– mitochondrial dysfunction exacerbation  
vitamins (304−306) – cellular metabolism regulation – vitamin-associated diseases
– immunomodulation – schizophrenia
– cell proliferation – autism
– vitamins supply – dementia
– IBS
– IBD
neurotransmitters (307−309) – gut motility regulation – Parkinson’s disease
– memory support – autism spectrum disorder
– stress response – IBD
– nervous system – IBS
– immune response
lipids (184,310,311) – systemic inflammation promotion – diabetes
– hyperinsulinemia regulation – obesity
– immunomodulation – nonalcoholic fatty liver disease
– bile acid synthesis – hyperinsulinemia
– hypercholesterolemia
– chronic hepatitis C
gases (307,312−316) – gut motility – colitis
– gut inflammation – ulcer
– epithelial secretion – IBS
– mucosal blood flow

Gut Microbiota Metabolites’ Role in Digestive System

Gut microbiota imparts specific function in the host’s digestive system, in nutrient metabolism, xenobiotic and drug metabolism, in preservation of the integrity of the intestinal mucosal barrier, and in protection against pathogens (Figure 13).

Figure 13

Figure 13. Gut microbiome metabolite classes and their roles in digestive system functions.

Gut microbiota mostly get nutrients from dietary carbohydrates. Fermentation of the carbohydrates, including indigestible oligosaccharides by the microbes in the colon, such as Bacteroides, Roseburia, Bifidobacterium, Faecalibacterium, and Enterobacter, ends in the synthesis of SCFAs, such as butyric, propionic, and acetic acids, which are important sources of energy for the host. (275) Gut microbiota have a positive role in lipid metabolism, as well, by controlling the lipoprotein lipase activity inhibition in adipocytes. (31)
Intestinal microbiota also exhibit a resourceful protein metabolizing machinery, which operates by means of the microbial proteinases and peptidases in conjunction with the human proteinases. Examples include the conversion of l-histidine into histamine by the bacterial enzyme histamine decarboxylase and glutamate to γ-amino butyric acid by glutamate decarboxylases. (276,277)
SCFAs can regulate the pH value in the intestine and regulate the absorption of water, sodium, calcium, and magnesium. Furthermore, SCFAs, especially butyrate, provide more than 70% of the energy for the intestinal epithelial cells on top of their abilities to inhibit the multiplication and growth of pathogenic bacteria and the activity of intestinal inflammatory mediators, thus playing an anti-inflammatory role in the intestinal tract. (278)
Lipid metabolites can affect intestinal permeability and intestinal immunity. The gut microbiota can produce lipopolysaccharides that could stimulate proinflammatory mediators, thereby disrupting the body’s immune system and inducing local and systemic inflammatory responses. (279) Sphingolipids can be produced by the intestinal symbiotic bacteria Bacteroidetes and Prevotellaceae. It has been found in animal studies that sphingolipids can also aggravate intestinal inflammation. (280)
Indole-derived metabolites are produced by fermentation via Clostridium sporogenes and Escherichia coli. Such metabolites are able to participate in the regulation of gastrointestinal disorders by influencing the gut–brain axis and protecting against stress-induced damage in the gastrointestinal tract. Tryptophan is a key monoamine neurotransmitter involved in the regulation of central neurotransmission and intestinal physiological functions, and studies have shown that the gastrointestinal microbiome can regulate the gut–brain axis through tryptophan metabolism. (281,282)
Gases can be produced by gut microbiota as a result of the fermentation process. These gases include hydrogen (H2), methane (CH4), carbon dioxide (CO2), hydrogen sulfide (H2S), and nitric oxide (NO), which can modulate the gastrointestinal physiology of hosts. (236)
Xiao et al. (283) in a recent review article highlighted the important microbial metabolites in the context of host physiology in patients with different IBS subtypes. The abundance of microorganisms and their corresponding metabolites in constipation-predominant IBS (IBS-C) and diarrhea-predominant IBS (IBS-D) differ, thereby providing a new avenue for the diagnosis and treatment of different IBS subtypes in the future. These microbiota-derived metabolites, such as bile acids (BAs), SCFAs, vitamins, amino acids, 5-HT, and hypoxanthine, can be produced directly by bacteria, or from dietary or relevant substrates. Fluctuations and alterations in the levels of metabolites produced by the host or microbiota provide insights into their interactions during IBS. Moreover, low levels of hypoxanthine may be associated with colonic epithelial energy and capacity for mucosal repair with hypoxia. Purine starvation has been identified as a potential novel mechanism underlying IBS with lower fecal hypoxanthine abundance in IBS-C and IBS-D. Additionally, mucosal biofilms are an endoscopic feature of IBS and are associated with bacterial and BA metabolites dysbiosis. Additionally, deficiency in levels of both vitamins D and B6 have emerged as causative factors in IBS symptoms pathogenesis. (283)
Microbial dysbiosis and metabolites derived from interaction of the host and gut microbiota have been reported as an intermediate link contributing to the development of functional constipation via various signal pathways, including but not limited to SCFAs, BAs, and methane that occupied a more important position. (153) 5-HT is also involved in the modulation of gut motility and secretory, as well as sensory, transmission in patients with constipation. (284) Altogether, current studies have provided us with a new conception on the microbial mechanisms and therapeutic targets of constipation.
The gut microbiome metabolites and their function and associated digestive diseases/disorders are summarized in Table 3.

Gut Microbiome–Brain Axis

ARTICLE SECTIONS
Jump To

CNS and the human GI tract communicate through the gut–brain axis (GBA). This bidirectional connection involves neuronal, endocrine, and immunological mechanisms. The gut is considered as our “second brain,” because of its hosting the enteric nervous system (ENS), a neural network that allows the gut to work without instructions from the brain. The ENS maintains control of our digestive system; it plays an important role in peristalsis, secretion, and pain perception. There is mounting data that gut microbiota are the source of a number of neuroactive and immunocompetent substances, as shown above, that help to shape the structure and function of brain regions involved in the control of emotions, cognition, and physical activity and contribute to the proper maintenance of gastrointestinal homeostasis. Most GI diseases are associated with altered transmission within the GBA that are influenced by both genetic and environmental factors. (317)
Functional gastrointestinal disorders (FGIDs) were previously considered as purely functional disorders with no scientific confirmation of a clear pathogenetic mechanism. According to Rome IV, the phenotype of FGIDs results from an altered transmission of nerve and biochemical signals within the gut microbiome–brain axis with mechanisms controlled by both genetic and environmental factors. Consequently, FGIDs were recently renamed into disorders of gut–brain interactions. (318) The overlap of DGBI and CNS disorders has been documented, and it has been demonstrated that approximately one-third of IBS patients suffer from depression. It is estimated that psychiatric symptoms occur in at least 36.5% of FGIDs patients. Stasi et al. found that the highest prevalence of mental or spectrum disorders is in patients with functional constipation (60%) compared with patients diagnosed with FD (52.4%), IBS (36.5), and/or functional bloating (47.6%). The most prevalent psychiatric disorders observed in FGIDs were general anxiety disorder and panic. (319)
Recent advances in this field have enabled us to better understand some of the pathophysiological consequences of an aberrant reciprocal gut–brain network, including exacerbated gut inflammation disorders, altered responses to stress, as well as altered behavioral states. Therefore, the GBA presents an attractive target for the development of novel therapeutics for an ever-growing list of disorders related to mental and digestive health. Improved targeting of the gut microbiome–brain axis, for example through application of biotics, is expected to pave the way for the development of novel disease therapies and self-care products to promote and maintain heathy status. (320)

Irritable Bowel Syndrome

IBS is one of the most common DGBI worldwide and typically presents in early adulthood with symptoms including abdominal pain, bloating, and altered bowel habits. On the basis of the bowel habits, IBS can be classified into four subtypes: constipation-predominant, diarrhea-predominant, mixed-type (IBS-M), and undefined IBS (IBS-U). Symptom intensity varies over time and between individuals, but IBS has been reported, in severe cases to affect quality of life as much as renal impairment or diabetes. (321) IBS represents up to 50% of all referrals to gastroenterologists with a prevalence rate of up to 11% globally. (322) The recent consensus view is that IBS results from abnormal gut–brain interactions. Recent epidemiological data has suggested that in individuals developing both IBS and psychological features, the former preceded the latter in two-thirds of cases, and the latter preceded the former in one-third. (323) IBS is associated with abnormalities of central pain processing but also increased gut permeability, mast cell activation, disordered motility, and dysbiosis.
A recent genome-wide analysis study for 53 400 IBS patients and 433 201 controls highlights shared genetic pathways between IBS and mood and anxiety disorders. The study identified and confirmed six genetic susceptibility loci for IBS, and four of them are associated with mood and anxiety disorders, thereby suggesting, for example, that shared pathogenic pathways rather than anxiety cause abdominal symptoms. (324)
5-HT signaling is one particular pathway of importance in IBS pathogenesis. It has been demonstrated that a functional GI tract involves 5-HT signaling between enterochromaffin cells acting as sensory transducers, and the majority of 5-HT is synthesized, stored, and released by these cells, which interact with intrinsic and extrinsic sensory nerve afferents in the mucosal layer of the gut. (320) 5-HT signaling controls many GI functions, including secretion; vasodilation; peristalsis; and sensory perception, such as pain and nausea. (325−327) Moreover, serotonergic function and tryptophan metabolism are known to be altered in IBS patients. (328−331)
IBS pathophysiology implicates altered gut microbiota composition, impaired intestinal mucosal integrity, and low-grade inflammation. In addition to pathways through the circulatory system, several of these factors may also trigger fluctuations in the activity of the ENS with subsequent effect on the brain. (332) Furthermore, the vagus nerve can be modulated by diet-responsive gut microbes and metabolites, such as short-chain fatty acids, or endocrine factors, enzymes, and neurotransmitters, such as serotonin, dopamine, acetylcholine, glutamate, γ-aminobutyric acid, and noradrenaline. (333−336) Each of these factors are potentially affected by alterations in microbiota composition and are involved in IBS pathology. (337)
Identifying a clear IBS microbial signature is not an easy task because of the heterogeneity of the healthy gut microbiota. However, multiple studies have shown differences in the gut microbiota between IBS and healthy controls. A recent systematic review showed that IBS patients have increased levels of the bacterial families Enterobacteriaceae, Lactobacillaceae, and Bacteroidales, whereas Bifidobacterium, Faecalibacterium, and Clostridiales were decreased compared with healthy controls. (122) On the contrary, Hugerth et al. recently reported no distinct microbiota signature of IBS in a random Swedish population of 3556 participants. (338) Interestingly, intestinal bacterial composition has been reported to be highly dependent on sample type and regional localization. Also, mucosa-associated bacterial composition of the sigmoid colon differs between patients with IBS and healthy controls. (339)
One of the most consistent findings in brain neuroimaging of IBS patients has been alterations in the structure and function of key regions of the somatosensory network, including the globus pallidus, putamen, and caudate, which composes the basal ganglia. (340) Increased gray matter density in the hypothalamus and decreased gray matter density in the prefrontal cortex have been reported in the IBS brain. (341) In rectal distention experiments, patients with IBS had a differential brain response in the pain matrix and default mode network. (342) IBS patients showed increased engagement of endogenous pain faciliatory pathways and decreased levels of the endogenous pain inhibitory mechanism in the brain regions associated with visceral afferent processing and emotional arousal, including the left dorsal anterior cingulate gyrus and the bilateral anterior insulae. (343) A meta-analysis of adult studies that evaluated brain response to rectal balloon distension by functional MRI (fMRI) reported differences between healthy control subjects and patients with IBS in these brain regions. (344) IBS patients with a history of abuse reported increased pain and anxiety with rectal distension accompanied by similar fMRI changes. (345) The stress and arousal circuit demonstrated in human subjects by fMRI shares significant homology with the stress circuit related to CRF–CRF1 receptor signaling in rodents, thereby potentially implicating the HPA axis as a facilitator of gut–brain axis communication. (346)
Recently, evidence for disrupted subcortical and cortical regions mediated by gut microbial modulation has been emerging in IBS. An association between brain region-to-region functional connectivity and microbiota has been reported. Labus et al. found a correlation between Clostridia and Bacteroidia with connectivity of the thalamus, the basal ganglia (caudate nucleus, putamen, pallidum, nucleus accumbens), the superior part of the precentral gyrus, the anterior insula, and the ventral prefrontal regions in IBS patients. (347) Recently, the same group also reported on fecal metabolites and resting state fMRI where the differences in histidine, cysteine, glycine, glutamate, spermidine, and anserine were significantly associated with the alteration in the left dorsal part of the posterior cingulate gyrus to the left putamen. Also, the changes in histidine, tryptophan, uracil, 2-deoxyuridine, thymidine, and succinate were differentially associated with the alteration in the right superior frontal gyrus to the right putamen. Interestingly, this interaction may be mediated by aberrant tryptophan signaling in IBS, which is important because it is a substrate for serotonin synthesis. (348)
Previous studies have compared brain differences between IBS-C and IBS-D. These studies have examined task states and have shown group-related differences in brain networks involved in integrating emotions [emotional arousal network, (EAN)], perception [sensorimotor network, (SMN); salience network, (SAL)], visceral functions [central autonomic network, (CAN)], and pain processing [default mode network, (DMN); central executive network, (CEN); SMN; SAL; EAN; and others). (349) Prior studies comparing IBS-C, IBS-D, and healthy controls (HCs) undergoing aversive rectal stimuli have identified abnormal connectivity in the SAL (350,351) and EAN (343,350) in IBS-C and in the occipital network (OCC) (351) in IBS-D. This may indicate a greater importance of alterations in sensory, emotional, and autonomic responses associated with the perception of visceral pain and discomfort in IBS-C. A recent study has tested the hypothesis that IBS-C exhibits bowel-habit-specific changes in the brain that reflect altered sensory and emotional regulation processing of visceral inputs from the “top-down,” while IBS-D would have widespread gut microbiome and metabolome changes (e.g., tryptophan, SCFAs), which may translate to “bottom-up” brain changes (e.g., SMN, DMN). (349) Indeed, in IBS-D, the study’s findings showed a correlation between high levels of gut metabolites tryptophan and phenylalanine and aberrant connectivity in brain regions involved in processing unpleasant visceral stimuli (SMN) and self-related thoughts (DMN). (349) These results suggest that increased tryptophan in the gut may lead to loosened stool, and tryptophan-related signaling may travel to the posterior insula and increase pain perception and emotional salience in IBS-D, thereby suggesting a “bottom-up” signaling direction. However, IBS-C’s microbiome and metabolome resembled HC, and the increased connectivity in the default mode (DMN) and salience (SAL) networks compared with IBS-D may indicate abnormalities in the emotional physiological processing of visceral signals. (349) IBS-C’s relatively isolated brain changes may indicate a more “top-down” mechanism to produce the constipation-predominant phenome. That study by Sarnoff et al. has shown a link between the chronicity of IBS symptoms, B. stercoris and F. prausnitzii, and brain connectivity in the caudate nuclei.
Thus, we might be in the mere beginning of understanding how alterations in gut microbiota may lead to the disruption of the intricate host–gut–microbiota interaction: is it a cause or a result of IBS pathology? In the past decade, much knowledge has been gained from clinical microbiota-altering interventions, such as the low-FODMAP diet and fecal microbiota transplantation (FMT), which have emerged as debatably successful treatment strategies. However, their effects on the gut microbiome–brain axis are still far from understood. It has been proposed that probiotic amelioration of IBS symptoms may be acting indirectly through an anti-inflammatory mechanism. (352) Such anti-inflammatory mechanisms may also be partially responsible for the positive effects of dietary restrictions, such as those seen in the low-FODMAP (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) diet in IBS. (353−355) An alternate method for IBS treatment involves FMT from non-IBS individuals to patients with IBS. (356−359)

Functional Dyspepsia

Functional dyspepsia (FD) is a worldwide prevalent DGBI affecting 10%–30% of adults and 3.5%–27% of children worldwide. (360) The main clinical symptoms of patients are early satiety, postprandial discomfort, epigastric pain, epigastric distension, epigastric burning, loss of appetite, belching, nausea, and vomiting, which are often accompanied by anxiety and depression. (361) FD pathogenesis is linked to GI dysmotility, visceral hypersensitivity, impaired gastric tolerance, disrupted gastrointestinal mucosal integrity, abnormal function of the gut microbiome–brain axis, increased eosinophils in duodenum, dysbiosis, Helicobacter-pylori infection, postgastrointestinal infection, diet, genetics, and mental and psychological factors. (278)
An increasing number of studies have confirmed the close association between disturbance in the relative abundance and composition of the gastrointestinal microbiota and the occurrence and progression of FD. Actinomyces, Atopobium, Leptotrichia, Prevotella, and Veilonella counts differ between FD and control patients. (362) The finding was preceded by an observation that, in FD patients, gut barrier integrity is impaired and expressed as lowered transepithelial resistance; diminished expression of proteins of tight junctions; and lastly, elevated levels of mast cells, eosinophils, and interstitial lymphocytes. (363)
Surprisingly, FD patients not only had different gastrointestinal microbiota compared with non-FD, but also had different oral microbiota abundance and composition. Proteobacteria were the dominant bacteria in FD patients’ saliva, while Bacteroidetes were the dominant bacteria in healthy controls. The abundance of Spirochaetes in FD patients was higher than that in healthy controls, while the abundance of Fusobacteria, TM7, and Proteobacteria was lower than in healthy controls, and the levels of Kingella and Abiotrophia genus levels were also significantly different. (364)
Mental illness plays a significant role in the pathogenesis of FD. Anxiety at baseline has been shown to increase the risk of developing FD by almost 8 times after 10 years follow-up. (365) Interestingly, multiple studies highlighted that the prevalence of anxiety and depression is significantly increased in patients with FD compared with healthy people. Furthermore, pathophysiological research indicates that psychosocial factors and mental disorders may play a role in FD by modulating both visceral signal processing in the brain (366) and the effects of stress hormones on pain perception. (367) Furthermore, it is known that psychosocial factors and stress hormones also affect other aspects of the GI tract, such as motility, immune system activation, permeability, and microbiota.
However, FD symptoms are thought to induce anxiety or depression because of a cytokine response in low-grade intestinal inflammation, which plays an important role in the development of psychological distress in patients with FD. (368) A growing body of evidence suggests that the gut microbiota communicates with the central nervous system, possibly neuro-immuno-humoral pathways, thereby influencing brain function. (369) Furthermore, microbiota release neuroactive compounds, such as GABA, serotonin, dopamine, and acetylcholine, thereby acting locally on the enteric nervous system. Some of these neuroactive substances access the brain through the blood and the circumventricular organs or via the vagus nerve. Therefore, it could be hypothesized that a disturbed microbiome might affect mental health, followed by anxiety and depression. Thus, the mental disorders might be a consequence of the dysbiosis and, therefore, promote the development of FD, which may explain the findings that anxiety increases the risk of FD (365) and observations indicating that psychosocial factors and mental disorders may play a role in FD by modulating visceral signal processing in the brain. (366,370)

Functional Constipation

Functional constipation (FC) is a common DGBI with a global prevalence ranging approximately from 10.1 to 15.3% (371) and is characterized by difficult bowel movements and/or a sense of incomplete evacuation, thus influencing quality of life. (372) Previous studies showed that the gastrointestinal microbiota composition of constipation is clearly distinct from that of normal individuals. The species diversity of microbiota in the patient samples was lower than that in healthy subjects; it was also accompanied by significantly reduced levels of Bifidobacterium and Lactobacillus and an increased abundance of Desulfovibrionaceae. (373) Levels of butyrate-producing bacteria, such as Faecalibacterium and Roseburia, were significantly reduced in patients with FC. (373) It has also been confirmed that the relative abundance of methanogenic bacteria is increased in patients with slow transit constipation relative to healthy subjects. (373,374) In another study, Chen et al. collected 3056 fecal amplicon sequence data from five research cohorts and used machine-learning methods to construct the constipation discriminant model. The model identified 15 top-ranking biomarkers, particularly inflammation-related pathogenic bacterial genera Serratia, Dorea, and Aeromonas. (375) A recent shotgun metagenomics study confirmed the results of previous studies by showing that the relative abundance of Roseburia intestinalis, a prominent butyrate-producing bacterium, was reduced in patients with constipation in comparison with healthy controls, and the microbiome corresponding to constipation was enriched for pathways implicated in methanogenesis. (376) In contrast, the microbiome of healthy individuals was characterized by high levels of genes associated with carbohydrate, fatty acid, and lipid metabolism. (150)
Notably, different intestinal sites harbor certain gut microbiomes, yet the majority of recent research has focused on the analysis of fecal-derived microbiota, which are accessible via noninvasive sampling methods. However, luminal microbiota is generally considered to be representative of the distal large intestinal content. The mucosa-associated microbiota, which live in more intimate contact with the host, cannot be fully replicated by fecal microbiota. (377) In patients with constipation, there is even less similarity between fecal- and mucosa-associated microbiota compared with healthy controls and patients with diarrhea. (116) These differences may be due to drier stool allowing fewer signaling molecules to enter the mucosa (378) or the longer transit time providing more opportunities for the communities to diverge. (116) Hence, mucosa-associated microbiota are more likely to affect the host’s epithelial and mucosal function than luminal microbiota. (379) Comparative analyses between fecal and mucosal microbiota showed that the colonic mucosal microbiota composition was correlated with constipation (and was accompanied by a significant increase in Bacteroidetes), while the fecal microbial communities were correlated with colonic transit and methane production rather than constipation. (380) However, more evidence is needed to prove the relationship between the mucosal profile and constipation.
Slow gut transit has been associated with reduced fecal water content, higher fecal pH, higher microbial cell density and diversity, and a shift in microbial metabolism from saccharolysis toward proteolysis, as reflected by reduced levels of short-chain fatty acids and increased levels of branched-chain fatty acids (BCFA). (381) It is likely that once easily accessible carbohydrate sources become scarce in the colon, the gut microbes switch to ferment dietary and mucin-derived proteins. While saccharolysis by the gut microbiota gives rise to SCFA that are beneficial for the host and a source of energy for the colonocytes, proteolysis can lead to the accumulation of compounds such as BCFA, phenols, indoles, ammonium (NH3) and hydrogen sulfide (H2S) that are generally considered detrimental for health. Moreover, hydrogen (H2) with carbon dioxide (CO2) or formate can be converted into methane (CH4) by methanogenic archaea, which are also linked to slower transit time. In addition, the production and circulation of secondary bile acids and hydrolysis of host-derived glucuronides excreted via bile can also be affected by alterations in gut transit time. (381)
FC in patients would accompany mental disorders like anxiety and varying severity of depression. (382) Evidence from recent neuroimaging studies illustrated that FC patients had significant structural and functional alterations in brain regions that are involved in visceral sensorimotor, cognitive control, and emotional regulation, (383−389) thereby confirming its reclassification as DGBI. Among these altered brain regions in FC patients is the anterior insula, which is generally regarded as a critical node for its essential role in processing interoceptive signals, modulating visceral activities, and regulating emotions and cognitions. (383,385,389−391)

Stress and Stress Resilience

Chronic stress is rapidly becoming a global societal challenge. Stress constitutes a state of threatened homeostasis triggered by intrinsic or extrinsic adverse forces (stressors) and is counteracted by an intricate repertoire of physiological and behavioral responses aiming to maintain/reestablish the optimal body equilibrium (eustasis). Stress is a nonspecific response of the body to any demand imposed upon it that disrupts the body homeostasis and manifests with symptoms such as anxiety, depression, or even headache. (392) Stress can be hardly avoided in the present-day, modern, competitive life. Although eustress is important for people’s rapid reaction to threats, chronic stress is associated with detrimental effects on physical health and adverse implications on the immune, neuroendocrine, and central nervous systems. (392)
Acute stress activates the HPA axis, thereby resulting in an immediate release of cortisol. This response prepares the individual to defend against or escape from a threat. After the threat subsides, normal homeostasis should return. However, when that fails to occur, chronic activation of the stress response results in dysregulation of the HPA axis and an increased risk of subsequent diseases/disorders. (320) It also acts on the gut to increase the release of proinflammatory mediators, which leads to increased gut permeability. (393) Repeated exposure to stress can initiate a vicious cycle of low-grade inflammation and negatively impacts the intestinal barrier and immune signaling within the gut. (394,395)
A link between stress and the abundance of lactobacilli in mice was discovered for the first time more than 40 years ago. (396) Several preclinical studies have documented that stress impacts gut microbial composition in a number of different hosts using different stress models ranging from water avoidance to maternal separation, heat, and acoustic stress and overcrowding. (320) These results have shown clinical relevance and translated into human studies, thereby showing the influence of stress on gut microbiota and gut microbiota on stress modulation through different stressors, such as surgical intervention, academic examination, or military training, among others. (397−400)
Maternal stress during pregnancy displays a distinct fecal microbiota profile, which has generational consequences. The maternal microbiota influences offspring microbiota and correlates with hyper-reactivity of the HPA axis, together with other perinatal factors, as a key determinant of offspring outcomes. These findings have been confirmed in humans in a population-based study whereby infants born to mothers with high cumulative stress during pregnancy exhibited an aberrant microbial composition. (401)
The effects of early life stress on the microbiota may extend to adulthood. (49) It is, therefore, plausible that changes in the gut microbiota due to stress at least partially mediate the onset of stress-related depressive or anxious episodes. Correlational studies have shown that fecal microbiota composition in individuals with anxiety or depression differs from that in healthy controls. (202,402,403) Women with a higher fecal Prevotella abundance experienced increased negative emotional response to viewing negative images and lower brain activity in the hippocampus than those with a higher Bacteroides abundance. (404)
Stress resilience is the ability to experience stressful events without the development of chronic elevated stress (psychological and/or biological) and associated changes in emotional behavior. (405,406) Stress susceptibility is related to psychological factors, such as passive coping skills and high emotional reactivity, but is also associated with biological factors such as hypo- or hyper-responsiveness of the stress response system, sex hormones, central and peripheral immune activation, and glucocorticoid resistance. (407) The gut microbiome is a biological factor that is emerging as a possible influencer of stress resilience. The broad influence of the gut microbiota on human health, including psychiatric health, has begun to be realized and understood over the past decade. Preclinical studies have reinforced this principle showing a connection between the gut microbiome–brain axis and stress resilience. Li et al. recently reported that certain mice exposed to chronic stress were found to be resilient to stress-induced corticosterone and anxiety-like behavior. These mice contained a relative abundance of Lactobacillus species within their gut microbiome. Subsequent stress-susceptible mice saw decreased anxiety-like behavior and corticosterone levels with Lactobacillus murinus supplementation. (408)
Modulation of the gut microbiome has emerged as a possible way to improve stress resilience and mental health. In 2013, Dinan et al. coined the term psychobiotics, which refers to live microorganisms when ingested in adequate amounts that produce a health benefit in patients suffering from psychiatric illness; the definition has been expanded to include other interventions that modulate the gut microbiome, such as prebiotic. (409) The term psychobiotics has since been widely adopted by neuroscientists conducting research on neurodegenerative diseases and depressive disorders in order to describe the use of different biotics to tackle depression, stress, anxiety, and other mental health complaints through the GBA.

Sleep

Adequate sleep quality and sufficient duration are necessary to support both mental and physical health and overall quality of life. (410) Inadequate sleep in either duration and/or quality has been increasingly recognized as a global public health issue. Sleep disturbances are typically characterized by a decrease in one’s ability to initiate and maintain sleep and by a reduced proportion of the deeper, more restorative sleep. (411) Increased risk of developing chronic diseases, such as obesity, type 2 diabetes, heart disease, some types of cancer and mental illness, has been associated with inadequate sleep. (411,412)
Evolving evidence has shown the impact of gut microbiota on sleep. In humans, previous research has shown that partial sleep deprivation can alter the gut microbiome composition in as little as 48 h; (413) however, longer periods of sleep deprivation apparently do not have this effect. (414) A more recent study showed that high sleep quality was associated with a gut microbiome containing a high proportion of bacteria from the Verrucomicrobia and Lentisphaerae phyla and that this was associated with improved performance on cognitive tasks. (415)
Microbiome diversity was positively correlated with sleep efficiency and total sleep time and was negatively correlated with the sleep fragmentation, thereby indicating that diversity of the gut microbiome promotes healthier sleep. However, two previous studies in humans suggested that microbiome diversity is insignificantly affected following a period of sleep restriction. (413,414) A critical difference between these studies is that the former study measured sleep over an extended period of time (one month), while the latter two studies manipulated sleep by experimentally restricting sleep. Accordingly, it is possible that short-term manipulations to sleep do not influence the gut microbiome diversity, but rather that microbiome diversity can influence sleep in the long term.
Gut microbiota may affect sleep status via degradation products, such as muramyl peptides (MPs), lipopolysaccharide, and melatonin. (416) These degradation products could activate immune cells that lead to the release of cytokines, which could affect sleep. Cytokines represent a potential critical interface between sleep physiology and gut microbiome composition. The acute phase pathway, cytokines IL-1β and IL-6, in particular, are strongly associated with sleep physiology. IL-1β is a major somnogenic factor. (416) IL-1β administration in human and nonhuman animals increases spontaneous sleep and fatigue, and IL-1β increases with ongoing sleep loss. (416) Unlike IL-1β, IL-6 is not a direct somnogenic factor, but sleep loss results in increased IL-6 levels. (417) In the gut, IL-6 and IL-1β-mediated inflammation fluctuates in response to stress and disease. (418) For example, intestinal mucositis results in an increased expression of IL-6 and-IL-1β in the small intestine (419) and in serum and colon tissue (420) in mice. In humans, chronic stress, alone, increases IL-6 and-IL-1β.
Alterations in the microbiome have been shown to influence neurotransmission of serotonin in both the peripheral and central nervous system. (411) While this may convey a positive impact on mood and psychological well-being, (421) it also has the potential to influence sleep (422) as serotonin is acetylated and, then, methylated to yield melatonin─the hormone important in helping regulate sleep/wake cycles. (423)
A recent meta-analysis involving 36 studies showed that sleep disorders were common in IBS, and the prevalence rate was 37.6% (95% confidence interval: 31.4% to 44.3%). (424) The pooled odd ratio revealed that sleep disorders were significantly associated with IBS. The reason why sleep disorders are associated with IBS remains unclear; however, the gut microbiome–brain axis could play an important role in the pathogenesis of both. Modification of the autonomic nervous system activity has been observed in cases of sleep deprivation, which indicates that sleep disorder might be associated with autonomic dysregulation. (425,426) It has been postulated that sleep inhibits the HPA axis, and sleep disorder may result in a 24 h increased secretion of cortisol. (427) Moreover, IBS symptoms, such as abdominal pain, may activate the sympathetic nervous system and, hence, reduce sleep efficiency. (428) Microbiome modulation has been shown to influence melatonin production and modulate IBS symptoms in individuals with a normal circadian rhythm. (429) Overall, although the reason for sleep disorders seen commonly among IBS patients is obscure, a gut microbiome–brain axis disorder may underlie this association. (427)
An additional layer of evidence has been shown by the intertwined interactions between the gut microbiome and the central and peripheral circadian rhythms. (430) The disruption of the host circadian rhythm alters the gut microbiome equilibrium. In addition, the microbiome is able to mediate host clock gene expression in peripheral organs and the suprachiasmatic nucleus.
Intestinal bacteria have shown inherent circadian rhythms, as shown in previous metagenomic studies. (430) Diurnal fluctuations in abundance and activity have been observed in Clostridiales, Lactobacillales, and Bacteroidales, which account for ∼60% of the microbiota. (431) Studies on human stool samples of Enterobacter aerogenes have demonstrated responsivity to the circadian hormone melatonin, as well as a daily rhythm. (432) The gut epithelium experiences differential bacterial species and metabolites depending on the time of day and expresses toll-like receptors that sense microbiotal metabolites in a rhythmic pattern. (433) It has been proposed that the gut microbiome influences the rhythmic expression of the host’s internal clock by signaling molecules, such as butyrate, and by oscillations in microbiotal bacterial content in response to feeding patterns. (434)

Cognitive Function

Abundant evidence supporting the role of the gut microbiota in modulating cognitive function is mostly based on animal research. However, few studies have examined the influence of gut microbes on human cognition and supported the clinical relevance. One of these studies showed that the gut microbiota composition of obese and nonobese subjects was linked with scores in speed, attention, and cognitive flexibility coupled with alterations in neural activity in the thalamus, hypothalamus, and amygdala, thereby suggesting that obesity affects the microbiota composition and subsequent cognitive performance. (435) Additionally, the microbiota composition in 1-year-old babies was associated with cognitive development. Three groups of microbial composition have been identified where better performance was seen in the group with higher levels of Bacteroides. (436) This group was also less likely to be born via C-section, which supports the previous observation linking delivery mode with child cognitive development, (437) thereby highlighting the importance of gut microbiota colonization in cognitive development and function. (436)
Microbiome modulation has demonstrated beneficial effects on cognitive performance. Lactobacillus strains have improved cognitive performance in healthy elderly subjects. (438) Fermented milk product supplemented with a probiotic has been shown to modulate the activity of brain regions involved in cognitive performance during an emotional attention test in healthy women. (439) Also, the modulation of the microbiome via inulin prebiotic has been shown to improve memory and mood in healthy individuals. (440)
These results taken together indicate the potential role of the gut microbiome–brain axis in regulating cognitive performance and that microbiome modulation could be a promising approach for improving cognitive function in both healthy and vulnerable individuals. However, much more work is needed to understand why specific microbiome-related interventions have the potential to modulate cognition.

Emotional Well-being

Gut microbiota is emerging as a key mechanism for modulating emotional well-being. (441) In fact, emotional disorders, such as depression and anxiety, are frequently accompanied by functional gastrointestinal disorders, which suggests an association between gut function and psychiatric diseases. (442,443) Recent research reveals the gut–brain axis association with the vagus nerve plays an important role in emotional well-being. The subdiaphragmatic vagus nerve is a major modulatory pathway between the brain and gut microbiota. (444) Data suggest that fecal microbiota from depressed mice produce depression-like phenotypes and abnormal gut microbiome composition when transplanted to nondepressed mice via the subdiaphragmatic vagus nerve. (445)
Gut microbiota is emerging as a key mechanism for the modulation of emotional well-being. (441) In fact, emotional disorders, such as depression and anxiety, are frequently accompanied by functional gastrointestinal disorders, which suggests an association between gut function and psychiatric diseases. (442,443) Findings from a longitudinal study further suggest that intestinal infections significantly predict the future onset of anxiety disorder. (446)
Evolving clinical evidence indicates the significant links between the gut and emotion; for example, altered gut microbiota composition (447) was reported in patients with depressive disorder in terms of fecal microbial diversity, as well as the level of the genus Faecalibacterium. (403) In healthy adults, self-rated higher quality of life and favorable personality types (high in openness and conscientiousness) were associated with the composition of certain gut microbiota (i.e., Faecalibacterium, Coprococcus, and Lachnospiraceae), as well as an enriched diversity of the gut microbiota community. (448,449)
Recent studies suggest that enterotypes play a role in regulating the association between the gut microbiome and mental health. (404,449) Enterotypes refer to robust stratified clusters on the basis of the variation found in the levels of one of three genera in the gut: Bacteroides (enterotype 1), Prevotella (enterotype 2), and Ruminococcus (enterotype 3). (450) Individuals’ enterotype cluster depends on, in part, long-term diet─i.e., the amount of ingested animal protein/saturated fats (Bacteroides-type) versus carbohydrates/simple sugars (Prevotella-type)─whereas they are less influenced by the hosts’ body mass, age, and sex. (450−452)
A recent brain imaging study found distinct patterns of the emotional process and brain connectivity between stratified enterotypes; clusters with a greater abundance of Prevotella show higher levels of emotional response, along with prominence in the connectivity of emotional, attentional, and sensory-processing brain regions when compared with Bacteroides-dominant clusters. (404) A large-scale microbiome study also revealed that the Bacteroides-enriched enterotype is significantly associated with a lower score on the subjective feeling of quality of life, as well as a higher score regarding depressive symptoms.
A recent exploratory study revealed that gut microbiome diversity is related to emotional well-being and that enterotypes significantly moderate the links between emotional well-being and gut microbiome diversity. (441) The enterotypes did not alter mood status, itself, but moderated the strength of the association between one’s mood and gut microbiome diversity. In the Prevotella-dominant group, emotional status was more closely related to gut microbiota diversity, such that a positive effect was associated with increased gut microbiota diversity. However, in the Bacteroides-dominant group, one’s mood status was not significantly associated with gut microbiome diversity. This finding is in line with the results of Tillisch et al. (404) in that only the high-Prevotella group displayed an increased response to affective images in the limbic system. Such findings suggest a significantly tighter connection between emotional well-being and the well-being of the gut microbiota community, particularly in the Prevotella-dominant condition.
There is an evolving body of evidence suggesting that the modulation of the gut microbiome by biotics administration or via food supplements (i.e., psychobiotics) may closely affect one’s mood. Benton et al. (421) showed that the consumption of probiotic-containing yogurt improved the self-reported mood of those whose mood was initially poor. Messaoudi et al. (176) showed that consumption of the probiotics reduced anxiety and depression scores in subjects with reduced urinary free cortisol. Also, consumption of Lactobacillus helveticas and Bifidobacterium longum reduced somatization, depression, anger and hostility, hospital anxiety, depression-scale global scores, and self-blame scores on coping checklists and increased focus on problem solving, but there was no effect on perceived stress. (176) Moreover, the administration of a combination of Lactobacillus acidophilus, Lactobacillus casei, and Bifidobacterium bifidum for 8 weeks improved the depression score. (453) In a 2017 systematic review by Wallace and Milev of 10 clinical trials, most of the studies found positive results on measures of depressive symptoms. (454) A recent meta-analysis (455) included 30 randomized placebo-controlled studies and revealed that most probiotics did not affect mood, anxiety, depression, and psychiatric distress when compared with placebo at the qualitative questionnaire level; however, on the quantitative meta-analysis level, probiotics intervention showed slightly significant effect compared with placebo. In addition, EEG and imaging studies summarized in a recent review proved that probiotics are able to exert effects on CNS function in humans; although, the number of studies is still low. (455)
Overall, there is emerging evidence on the link between one’s emotional status and gut microbiome diversity and composition. The current evidence suggests that emotional well-being and the feeling of happiness can be associated with gut microbiota profiles in healthy adults, especially when stratified by enterotype. The enterotype-specific links between emotional well-being and gut microbiome diversity suggest that enterotypes may work as an individually tailored intervention. With the expanding interest in the role of the gut microbiome–brain axis in mental health, this nascent field still needs to build up empirical evidence to fill the gap in our understanding of how gut microbiota communicates with the brain to affect emotional well-being and the feeling of happiness.

Prebiotics, Probiotics, Synbiotics, Postbiotics, and Psychobiotics

Probiotics

Probiotics are defined by the International Scientific Association for Probiotics and Prebiotics (ISAPP) as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host. (93,456) Probiotics usually comprise bacteria, including, among others, Lactobacillus, Bifidobacterium, and Bacillus, although a few strains of the yeast Saccharomyces have also been included in probiotic cultures. Probiotic microorganisms must have several features, including a demonstrated benefit to the host and the recognition that they are safe for human consumption, i.e., they have generally been recognized as a safe designation. They are resistant to acid and bile salts that are encountered in the GI tract. The ability to adhere to the intestinal epithelium is a useful trait that promotes both the persistence of the probiotic and host interactions. Other useful features are that they are easily cultured and are resistant to drying, freezing, and freeze-drying so that the probiotic may be produced and stored in bulk. Probiotics can enhance immune function, promote fiber assimilation for SCFA production, and help suppress pathogens in the GI tract. (93,457,458)

Prebiotics

Prebiotics are defined by ISAPP as a substrate that is selectively utilized by host microorganisms conferring a health benefit. (31,93) Therefore, such a definition expands beyond the classical prebiotics composed of polysaccharide carbohydrates and could include any other nondigestible microbiome-fermentable ingredients, such as herbal secondary metabolites and flavobiotics. The definition stresses that the effects must be microbiota-mediated and that the beneficial health effects must be documented for a substance to be considered a prebiotic. (459) The most well-known products of microbiome-derived fermentation are SCFAs, such as butyric acid, acetic acid, or propionic acid, which have a beneficial effect on the host. Moreover, prebiotics modulate lipid metabolism, increase calcium absorption, have a positive effect on the immune system, and reduce the risk of broad diseases. (458)

Synbiotics

ISAPP defined synbiotic as a mixture comprising live microorganisms and substrate(s) selectively used by host microorganisms that confer a health benefit on the host. The panel concluded that defining synbiotics as simply mixtures of probiotics and prebiotics could suppress the innovation of synbiotics that are designed to function cooperatively. The ISAPP panel differentiated synbiotics into two groups: complementary synbiotics and synergistic synbiotics. A complementary synbiotic that has not been designed so that its component parts function cooperatively must be composed of a probiotic plus a prebiotic. A synergistic synbiotic is a synbiotic for which the substrate is designed to be selectively utilized by the coadministered microorganisms. (460)

Postbiotics

Postbiotics are a class of products that has emerged in the last 10 years. Postbiotics are defined by the ISAPP as preparations of inanimate microorganisms and/or their components that confer a health benefit on the host. Effective postbiotics must contain inactivated microbial cells or cell components, with or without metabolites, that contribute to observed health benefits. (458,461) Postbiotics, because they are derived from probiotic microorganisms, have many of the same benefits as probiotics. The effects of postbiotics are often more reliable and predictable than probiotics. Postbiotics tend to have a longer shelf life than probiotics and they are more target-specific and safer in terms of their interaction with the human GI tract. (93)

Psychobiotics

The term psychobiotic was coined to describe bacteria that confer mental health benefits. Psychobiotics have demonstrated the ability to improve mood, reduce anxiety, and enhance cognitive function in both healthy populations and patient groups. While the term psychobiotics originally referred to beneficial live organisms, such as bacteria that are specifically beneficial for mental health, (409) the definition has been expanded in recent years to include prebiotics whose effect on the brain is bacteria-mediated. (462) It is also worthwhile considering a wider definition of psychobiotics to include any substance that exerts a microbiome-mediated psychological effect or at least possesses psychobiotic properties, such as probiotics, prebiotics, synbiotics, and postbiotics. (462,463) Recently a new term “phyto-psychobiotics” has been coined to describe medicinal plants whose mental effects are mediated via gut microbiota modulation by prebiotic-like effects, postbiotic-like effects mediated by the active secondary metabolites produced by the gut microbiome from the nondigestible herbal ingredients, or even by antibiotic-like effects as in the case with some medicinal herbs that have a mental impact by reducing the level of pathogenic bacteria. (464)

Pre-, Pro-, Postbiotics, and Fecal Microbiota Transplantation in the Development Pipelines

A search of the CAS Content Collection (84) reveals the top organizations for research and journal publications related to the gut microbiome and mental and digestive health. All these top players are universities and research institutes, and the University College Cork, the Chinese Academy of Science, the University of California, and McMaster University lead the field (Figure 14A). The lead universities and medical centers related to patenting activity for the gut microbiome in mental and gut health include the University of California, Johns Hopkins University, and the University of Texas (Figure 14B).

Figure 14

Figure 14. Top universities, research institutes, and hospitals publications (1967–2022) related to gut microbiome research in mental and gastrointestinal health: (A) journal publications and (B) patents.

Private Investment

Researching the overall global private investment activities of the microbiome field provides insight into the commercial interest into this area. Performing a search of prebiotics, probiotics, and the microbiome within PitchBook, an online source for investment data, reveals the overall venture capital activities. The search revealed that both capital raised and deal counts from venture capital investment are rising within this industry. (465) From 2014 through 2018, the total capital raised increased from $250 million to over $1 billion. Deal counts followed the same pattern from 2014 to 2018 and increased from 25 to 125. The number of deal counts in 2019 further increased; however, the overall capital raised fell to just under $900 million. Deal counts continued to rise for 2020 and 2021, along with capital raised, which totaled over $2.1 billion, while 2022 ended with a slight decrease in total venture capital investment (Figure 15). The venture capital investment data in this area clearly shows a recent and increasing commercial interest surrounding prebiotics, probiotics, and the microbiome, thereby revealing its potential promise for therapeutic applications.

Figure 15

Figure 15. Overall capital raised and deal counts of venture capital investment for the prebiotic, probiotic, and the microbiome field ($) (source: pitchbook.com).

Companies and Academic Institutions Investigating Treatment of Mental Disorders and DGBI through Gut Microbiome Modulation

With the GBA being a bidirectional communication network, we herein examine a highlighted selection of global companies and academic institutions researching and producing prebiotics, probiotics, postbiotics, and utilizing fecal microbiota transplantation to treat a variety of consumer health-related mental disorders and DGBI. This global analysis of worldwide companies and academic institutions, while extensive, is not comprehensive and provides insight into both the present and future state of the field.

Probiotics

Companies, along with universities and health institutions, are utilizing probiotics for the treatment of mental disorders at slightly different rates (Figure 16). Companies are focusing more on stress than universities and medical institutions, with all organizations researching anxiety at about the same rate. Universities and medical institutions are researching depression, cognition impairment, and sleep disorders at a higher rate than industry. With universities and medical institutions on the forefront of research, this shows the up-and-coming possibilities for probiotic products for the treatment of depression, cognition impairment, and sleep disorders being produced for consumers in the future. Research in the area of utilizing probiotics for DGBI is much more extensive and historically established (Supplemental Table 1) than mental health disorders (Figure 16). Companies, universities, and medical institutions research most DGBI at a similar rate, with universities and medical institutions having a higher focus on IBS. IBS is more prevalent among those who eat a Western diet. With more of the world’s population adopting a Western diet, the prevalence of IBS is expected to increase. (466) The research among universities and medical institutions is shadowing this trend as researchers evaluate probiotic treatment options for this DGBI increasing in prevalence (Figure 16).

Figure 16

Figure 16. (Left) The percentage and number of analyzed global organizations utilizing probiotics for mental disorders and DGBI treatment. (Right) The percentage and number of analyzed global organizations utilizing prebiotics for mental disorders and DGBI treatment.

Prebiotics

Fewer organizations are researching prebiotics for the treatment of mental disorders and DGBI when compared with probiotics (Figure 16). Prebiotics for DGBI are researched more than mental disorders, with more focus on IBS and functional constipation (Figure 16). This field has many future growth opportunities for both industry and research as it grows and becomes established. Similar to the number of organizations, the number of documents in the CAS Content Collections related to prebiotics is about three times lower compared with those related to probiotics. However, the growth rate in the prebiotics research has increased in the last two years.

Postbiotics

Postbiotics are the least researched therapeutic examined and are mainly limited to a small commercial presence (Figure 16). With such a small commercial and research presence, the opportunities are also abundant for postbiotic therapeutics.

Fecal Microbiota Transplantation

FMT research is found to be rare in comparison with other microbiome modulation strategies in mental disorders and newly established with only a very small number of companies and universities participating in this field (Figure 17). Most of the FMT research in DGBI focuses on IBS treatment (Figure 17). This field is currently showing clinical scientific evidence for the successful treatment of the serious and sometimes deadly condition of Clostridium difficile (C. Diff) infection. (467) Recently, Australia’s Therapeutic Goods Administration gave the world’s first regulatory approval to company BiomeBank for its biologic microbiome product called Biomictra for the treatment of C. Diff infection. (468) The US FDA followed a few weeks later with its official approval for the biologic drug consisting of live human-derived fecal microbiota, RBX2600 (Rebyota), produced by Ferring Pharmaceuticals, which is also for the treatment of C. Diff infection. (469) These first regulatory approvals open this method to endless opportunities for the treatment of many different diseases, and more studies are needed to prove its safety and efficacy.

Figure 17

Figure 17. (Left) The percentage and number of analyzed global organizations utilizing postbiotics for mental disorders and DGBI treatment. (Right) The percentage and number of analyzed global organizations utilizing fecal transplant for mental disorders and DGBI treatment.

Clinical Trials Landscape for Probiotics in Mental Disorders and DGBI

When examining clinical trials utilizing probiotics for the treatment of the mental disorders discussed within, there are currently a total of 52 clinical trials covering all stages between 2004 and 2022 listed on the US NIH clinical trials website. (470) The most studied mental health disorders are stress, followed by depression, anxiety, cognition impairment, and sleep disorders. Clinical trials utilizing probiotics for the treatment of DGBI are the most prolific and historically studied with 174 clinical trials. (470) The most studied DGBI are IBS followed by functional constipation, functional diarrhea, and functional dyspepsia.
The most researched probiotics for the treatment of mental disorders are the Lactobacillus species and a combination of Lactobacillus and Bifidobacterium species (Supplemental Table 1). For DGBI, the most widely used probiotic is the Lactobacillus species followed by the Bifidobacterium species, a combination of Lactobacillus and Bifidobacterium species, and finally the Saccharomyces species (471) (Supplemental Table 1).
Highlighted clinical trials examining probiotics as a treatment option for mental disorders and DGBI are explored in Tables 4 and 5. A select few are also examined in further detail below to showcase a variety of interventions and targeted conditions in clinical development, along with their status. While experimental data in this area is showing promising results, there are still conflicting results being reported.
Table 4. Highlighted Clinical Trials Utilizing Probiotics for the Treatment of Mental Health Disorders
clinical trial identifier condition intervention status
NCT05564767 (472) depression, anxiety, stress Bifidobacterium adolescentis Bif-038, Lacticaseibacillus rhamnosus LGG, Bifidobacterium BB-12 recruiting
NCT03494725 (477) stress, anxiety Lacticaseibacillus paracasei Lpc-37 complete
NCT04767997 (474) sleep disorder undisclosed probiotic formulation recruiting
NCT03601559 (478) cognitive impairment Lactobacillus paracasei Lpc-37 complete
NCT03615651 (479) stress, cognition impairment Lactobacillus helveticus, Bifidobacterium longum, Lactiplantibacillus plantarum complete
NCT05567653 (473) stress Lactobacillus helveticus Rosell-52, Bifidobacterium longum Rosell-175 recruiting
NCT03370458 (480) stress Lactobacillus plantarum DR7 complete
Table 5. Highlighted Clinical Trials Utilizing Probiotics for the Treatment of DGBI
clinical trial identifier condition probiotic intervention status
NCT02592200 (485) functional constipation Lactobacillus gasseri DSM 27123 complete
NCT04304170 (486) functional constipation Bifidobacterium animalis lactis (LMG P-28145) complete
NCT04662957 (487) diarrhea-predominant irritable bowel syndrome Bifidobacterium breve BB010, Bifidobacterium longum BL020, Bifidobacterium bifidum BF030, Bifidobacterium lactis BL040, Lactobacillus rhamnosus LR110, Lactobacillus paracasei LPC100, Lactobacillus acidophilus LA120, Lactobacillus casei LC130, Lactobacillus plantarum LP140, Streptococcus thermophilus ST25 complete
NCT05566171 (488) functional constipation Bifidobacterium lactis CNCM I-2494, Bifidobacterium lactis DN 173-010 enrolling by invitation
NCT01463293 (489) functional constipation Bifidobacterium lactis HN019 complete
NCT01102036 (490) functional constipation Lactobacillus paracasei F19, Lactobacillus paracasei LA-5, Bifidobacterium lactis BB-12 complete
NCT03721107 (491) diarrhea- and constipation-predominant irritable bowel syndrome Blautia hydrogenotrophica complete
NCT00534170 (492) functional diarrhea Lactobacillus casei Shirota, Bifidobacterium breve Yakult complete
NCT00794924 (493) functional diarrhea, functional constipation Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus delbrueckii subsp. bulgaricus complete
NCT02213172 (494) irritable bowel syndrome Bifidobacterium longum R0175, Lactobacillus paracasei HA-196 complete
NCT00807326 (495) functional diarrhea Saccharomyces boulardii complete
NCT05054309 (496) irritable bowel syndrome Bifidobacterium longum NCC3001 recruiting
NCT01099696 (497) functional dyspepsia Bifidobacterium infantis 35624 complete
NCT01887834 (498) irritable bowel syndrome Lactobacillus gasseri, Bifidobacterium bifidum, Bifidobacterium longum complete
NCT04605783 (499) functional diarrhea Saccharomyces boulardii CNCM I-745 not yet recruiting
NCT04950296 (500) irritable bowel syndrome with diarrhea Lactobacillus plantarum UALp-05 complete
NCT05149599 (501) irritable bowel syndrome Saccharomyces cerevisiae complete

Mental Disorders

Clinical trial number NCT05564767 is currently recruiting participants to assess the treatment of depression and anxiety in adults by utilizing probiotics Bifidobacterium alone and Lacticaseibacillus combined with Bifidobacterium. (472) Clinical trial number NCT05567653 is also recruiting for its study researching the treatment of stress in dancers with a probiotic product combination including Lactobacillus and Bifidobacterium. (473) Another study (NCT04767997) looking at the effects of probiotics on sleep disorders is also currently recruiting subjects. (474) Clinical trial number NCT03615651 researched the effect of a probiotic mixture containing Lactobacillus helveticus, Bifidobacterium longum, and Lactiplantibacillus plantarum on subjects’ functional brain responses during an emotionally stressful attention task. (472) Their findings showed a positive effect on brain responses in regions implicated in emotional and cognition processing, which supports the growing evidence that probiotics can help positively influence emotional regulation and brain function. (475) Another study (NCT04767997) that is looking at the effects of probiotics on sleep disorders is also currently recruiting subjects. Finally, a recently completed study (NCT03494725) found that supplementation with probiotic Lacticaseibacillus paracasei Lpc-37 significantly reduced perceived stress and anxiety in study participants. (476)

DGBI

Clinical trial number NCT05566171 is currently recruiting participants to research the treatment of functional constipation with two Bifidobacterium strains. Bifidobacterium was also researched in clinical trial number NCT04304170 and NCT01463293 for the treatment of functional constipation. Lactobaccillus and a combination of Bifidobacterium species and Lactobacillus were also researched for the treatment of functional constipation in completed clinical trials NCT01102036 and NCT02592200, respectively. Utilizing a combination of Streptococcus, Bifidobacterium, and Lactobacillus species, clinical trial number NCT00794924 researched the treatment of functional constipation and diarrhea in elderly hospitalized patients. The study showed positive results with the probiotics reducing days patients suffered from diarrhea or received laxatives, thereby displaying a positive effect on bowel movements. With diarrhea remaining a major public health concern in developing countries, study NCT00534170 researched the use of a probiotic drink containing both Lactobacillus and Bifidobacterium species for the treatment of diarrhea in young children. The study revealed the ingestion of a daily probiotic drink could prevent diarrhea in young children in a community setting within a developing country. (481) Another study (NCT00807326) also researched the treatment of functional diarrhea. They compared the treatment results of antigas/antidiarrheal drug combination loperamide/simeticone and probiotic yeast Saccharomyces boulardii. They discovered that while the probiotic did help alleviate symptoms, it was inferior to loperamide/simeticone. (482) Clinical trial number NCT01099696 also had discouraging results researching the treatment of functional dyspepsia with probiotic Bifidobacterium infantis 35624. While previous studies showed promise in patients with IBS, Bifidobacterium infantis 35624 did not show significant improvement in symptoms of abdominal discomfort and bloating in that study’s participants. Finally, a study (NCT04662957) researching a multistrain probiotic mixture of four Bifidobacterium, five Lactobacillus, and one Streptococcus species concluded its probiotic could offer benefits for patients with diarrhea-predominant IBS. (483) Another study (NCT03721107) showed more positive results for the treatment of both diarrhea and constipation-predominant IBS. Patients reported improvement in bowel symptoms and pain with the use of Blautia hydrogenotrophica. (484)

Clinical Trials Landscape for Prebiotics in Mental Disorders and DGBI

When examining clinical trials utilizing prebiotics for treatment of the mental disorders discussed within, there are currently a total of 15 clinical trials. (470) The most studied mental health disorder is stress, followed by anxiety, depression, sleep disorders, and cognition impairment. (470) A total of 50 clinical trials utilize prebiotics for the treatment of DGBI. (470) Similar to probiotics, the most studied DGBI using prebiotics is IBS, followed by functional constipation, functional diarrhea, and functional dyspepsia. (470) The most commonly used prebiotic in clinical trials is galacto-oligosaccharides (GOS), followed by other sugars such as fructan, glucan, and dextrose, along with various fibers.
Highlighted clinical trials examining the treatment of mental disorders and DGBI with prebiotics are explored in Tables 6 and 7. A select few are also examined in further detail below to showcase the variety of interventions and targeted conditions, along with their status in clinical development. The use of prebiotics for the relief of mental and gastrointestinal disorder symptoms is producing promising results.
Table 6. Highlighted Clinical Trials Utilizing Prebiotics for the Treatment of Mental Health Disorders
clinical trial identifier condition intervention status
NCT05372601 (504) stress GOS complete
NCT05239845 (505) sleep disorder polydextrose, GOS recruiting
NCT04324749 (506) cognitive impairment, stress roasted peanuts, peanut butter complete
NCT05528575 (507) stress GOS, inulin, resistant potato starch RS2 active
NCT04616937 (508) anxiety, cognitive impairment GOS complete
Table 7. Highlighted Clinical Trials Utilizing Prebiotics for the Treatment of DGBI
clinical trial identifier condition intervention status
ISRCTN54052375 (512) irritable bowel syndrome GOS complete
NCT04491734 (513) gastresophageal reflux maltosyl-isomaltooligosaccharides (MIMO) complete
NCT05207618 (514) irritable bowel syndrome with diarrhea chestnut and quebracho tannin extract complete
ACTRN12612001270808 (510) functional constipation green kiwi prebiotic, gold kiwi prebiotic complete
NCT05340712 (515) functional constipation infant formula with lactose (prebiotic) along with probiotics recruiting
Mental Disorders
Clinical study NCT04616937 researches the use of GOS for the treatment of anxiety by altering the gut microbiota. GOS increases probiotic bacteria Bifidobacterium abundance in the gut microbiome. The study reports that the supplementation of GOS may improve signs of anxiety and cognition impairment with an increase of reported attention. (502) Another recent study (NCT04324749) researched the prebiotic effect of peanut and peanut butter consumption on the cognitive and stress response of college students. The peanut prebiotic fiber and polyphenol content appears to enhance memory function and reduce stress because of the presence of both short-chain and very long-chain saturated fatty acids for healthy young subjects. (503)
DGBI
A published case study from 2019 showed a new observation that some patients taking a specific prebiotic soluble fiber, maltosyl-isomaltooligosaccharide (MIMO), had their gastroesophageal reflux symptoms resolve. (509) Clinical trial NCT04491734 followed about a year later to research this effect. The prebiotic MIMO reduced the severity and frequency of gastroesophageal reflux symptoms and improved the quality of life for participants. Clinical study ACTRN12612001270808 researched the use of a green kiwi prebiotic (inulin) and a gold kiwi prebiotic to treat functional constipation. The study showed a successful increase in bowel movements and also revealed that green kiwi prebiotic (inulin) supports the increase of two probiotic bacteria, Bifidobacteria and Lactobacillus, within the gut microbiome. (510) Another research study shows that gold kiwi prebiotic increased the abundance of Faecalibacterium prausnitzii within the gut microbiome, as well. (511) F. prausnitzii is a butyrate producer and displays anti-inflammatory effects. (511) When prebiotic GOS was tested in a clinical trial (ISRCTN54052375), it increased the probiotic bacteria Bifidobacteria within the gut. This helped alleviate symptoms of IBS, thereby showing that GOS is a potential therapeutic agent for this disorder. (512)

Clinical Trials Landscape for Postbiotics and FMT in Mental Disorders and DGBI

Postbiotic and FMT are the least researched among all clinical trials explored. When examining clinical trials utilizing postbiotics for the treatment of mental disorders, anxiety is the only disorder studied (Supplemental Table 1). Clinical trials utilizing postbiotics for the treatment of DGBI focus on IBS. (470) Only three clinical trials are researching FMT for the mental health disorders of depression, anxiety, and sleep disorders. Fecal microbiota transplants researching DGBI is higher with 27 trials researching both IBS and constipation.
Highlighted clinical trials examining the treatment of mental disorders and DGBI with postbiotics and FMT are explored in Tables 8 and 9. A select few are also examined in further detail below to showcase a variety of interventions and targeted conditions, along with their status in clinical development.
Table 8. Highlighted Clinical Trials Utilizing Postbiotics for the Treatment of Mental Disorders and DGBI
clinical trial identifier condition intervention status
NCT05475314 (517) irritable bowel syndrome microbially fermented postbiotic oat drink complete
NCT05562739 (518) anxiety multistrain postbiotic not yet recruiting
NCT05339243 (519) irritable bowel syndrome with diarrhea heat-treated Bifidobacterium longum ES1 recruiting
Table 9. Highlighted Clinical Trials Utilizing Fecal Microbiota Transplantation for the Treatment of Mental Disorders and DGBI
clinical trial identifier condition intervention status
NCT03822299 (522) irritable bowel syndrome fecal microbiota transplantation complete
NCT02092402 (523) irritable bowel syndrome fecal microbiota transplantation complete
NCT05035784 (524) functional constipation fecal microbiota transplantation recruiting
NCT05427331 (525) chronic insomnia fecal microbiota transplantation capsule recruiting

Postbiotic Clinical Trials Landscape for the Treatment of Mental Disorders and DGBI

Clinical study NCT05562739 (recruiting) is researching a multistrain postbiotic for anxiety treatment in individuals who were placebo nonresponders from part one of the trial. A recently completed study (NCT05475314) researched the use of a postbiotic fermented oat drink for the treatment of IBS. The study resulted in positive outcomes and showed symptom relief in IBS subjects. (516) Lastly, clinical trial NCT05339243 is currently recruiting for their study investigating both Bifidobacterium longum ES1 and the postbiotic heat-treated Bifidobacterium longum ES1 for the treatment of IBS symptoms in subjects with diarrhea-predominant IBS.

FMT Clinical Trials Landscape for the Treatment of Mental Disorders and DGBI

Clinical trial NCT05427331 is currently recruiting for its study examining sleep disorders. FMT through oral capsule administration will be researched to access sleep improvement in patients with insomnia. While fecal transplants are less prevalent for mental disorders, they have shown promise for DGBI with clinical trials by showing favorable results for the treatment of IBS. Clinical trial NCT02092402 showed effective treatment for patients with IBS when using a donor with a diverse microbial gut composition. (520) Another study (NCT03822299) also showed success for FMT in IBS. Just as with trial NCT02092402, it also found that the donor’s fecal composition with a favorable microbial signature and diverse gut microbiota was essential for successful treatment. (123) Three years after treatment, the study was still experiencing high response rates and long-standing effects. (521)

Noteworthy Probiotic and Prebiotic Patents

There are a diverse and growing number of patents related to probiotics and prebiotics in the CAS Content Collection. Listed in Table 10 are noteworthy prebiotic and probiotic patents related to the treatment of mental disorders and DGBI.
Table 10. Notable Prebiotic, Probiotic, and Postbiotic Patents
patent number title summary
WO2016085356A1 gold kiwifruit compositions and methods of preparation and use thereof Prebiotic compositions prepared from gold varieties of Actinidia chinensis. These prebiotic compositions treat or prevent DGBI, such as constipation, and IBS.
WO2022191767A1 GOS preconditioning L. reuteri and GOS in final formulation Enhancing the survival and activity of probiotic Lactobacillus reuteri strains by preconditioning L. reuteri with GOS. This method produces high synbiotic and beneficial effects of the probiotic bacteria in the gastrointestinal tract, such as boosting calcium and iron solubility, along with enhancing the production of lactic and acetic acid.
US20160058808A1 microbe-based modulation of serotonin biosynthesis Methods and probiotic compositions that can be used to modulate serotonin levels and adjust the composition of gut microbiota along with adjusting the level of serotonin-related metabolites.
WO2022182908A1 probiotic therapies for social deficit and stress response Bacterial species, including probiotic Enterococcus faecalis, for use in the treatment of social behavioral deficit symptoms, such as depression, by increasing social behavior and decreasing corticosterone levels along with c-Fos expression in the brain.
US9192618B2 method of treating constipation-predominant irritable bowel syndrome Prebiotic or probiotic agent that inhibits the growth of methanogenic bacteria or promotes the growth of competing intestinal microbiotia for the treatment of constipation predominant IBS.
US10022408B2 probiotic Bifidobacterium adolescentis strains Novel isolated strains of probiotic Bifidobacterium adolescentis for the prevention, alleviation of symptoms, or treatment of intestinal inflammatory conditions, such as IBS.
WO2005003329A1 novel GOS composition and the preparation thereof Novel strains of Bifidobacterium hifidum capable of producing a novel galactosidase enzyme activity that converts lactose to a novel mixture of GOS. The mixture of prebiotic oligosaccharides improves gut health by promoting the growth of bifidobacteria in the gut.
US20220040242A1 modulation of the gut microbiome to treat mental disorders or diseases of the central nervous system Methods of treating at least one symptom of a mental disorder and the central nervous system by modulating the amount of GABA produced in the gut. Also disclosed are methods of identifying and creating probiotic bacterial strains capable of producing GABA.
WO2016029198A1 process for the production of isomaltooligosaccharide Provides a method for the production of prebiotic oligosaccharides by the fermentation of dextransucrase-producing microorganisms.
WO2022214700A1 Lacticaseibacillus paracasei EM025-11 and uses thereof A probiotic strain of Lacticaseibacillus paracasei EM025-11 that adheres to intestinal epithelial cells and has anti-inflammatory activity by upregulating genes associated with immune engagement for the treatment of IBS with constipation.
US20220280576A1 Bifidobacterium longum and functional GI disorders Methods for treating functional GI disorders with probiotic Bifidobacterium longum ATCC BAA-999.
WO2019149941A1 postbiotic-based composition for the modulation of immune system activation and protection of mucosal barriers A fermented supernatant of Lactobacillus casei or paracasei species for the promotion of human health and prevention of inflammatory disorders. The postbiotic was shown to stimulate peripheral blood mononuclear cells and protect from endotoxic shock and Salmonella infection.
US8551498B2 solid composition containing Bacillus-type nonpathogenic bacterial spores Composition of spores of probiotic bacteria Bacillus useful in the pharmaceutical, veterinary, and nutrition fields.
ES2824536T3 use of microbial communities for human and animal health Mixture of probiotic bacteria belonging to at least six or seven bacterial species to prevent or treat GI disorders.
US20220233559A1 xylooligosaccharide as a multifunctional prebiotic Prebiotic mixture of xylooligosaccharides derived from sugar cane that were shown to modulate the levels of probiotic bacteria Bifidobacteria and Lactobacillus in the microbiome.
WO2022173764A1 nutritional plant-based foods and beverages, methods of manufacture, and methods of treatment Formulation of a prebiotic beverage whose ingestion modulates the gut microbiome by enhancing the growth of Bifidobacterium, improving the production of SCFAs, and reducing the levels Escherichia coli.
US20220315960A1 method for producing gamma-aminobutyric acid and fermented culture prepared thereby Process to produce gamma-aminobutyric acid from glutamic acid and a probiotic composition capable of this biotransformation. Ideally, the composition contains different probiotic bacteria Bifidobacterium and Lactobacillus strains.
WO2022208458A1 inactivated strains of bacteria, such as viable but nonculturable bacteria, compositions, and use thereof Postbiotic composition of different gamma-irradiated members of bacterial species Lactobacillus, Lacticaseibacillus, Bifidobacterium, and Lactiplantibacillus to treat several gastrointestinal disorders.
EP3932415A1 gut microbiota composition and uses thereof Probiotic composition for the prevention and/or treatment of a mental disorder with memory impairment by gut microbiome modulation for an increase in memory scores.

Conclusions and Perspective

ARTICLE SECTIONS
Jump To

Gut microbiota in humans evolving throughout life has been demonstrated to play a key role in health and disease. In healthy individuals, the intestinal microbiota has a multitude of beneficial functions, including metabolic energy utilization, protection from pathogenic attack, and immunomodulation. Furthermore, it is becoming increasingly documented that bidirectional signaling takes place between the gut and the brain and involves gut microbiota. This relationship encompasses various pathways, such as the vagus nerve; the hypothalamic–pituitary–adrenal axis; and immune, hormonal, and metabolic pathways to control various aspects of homeostasis, including the appropriate development and maintenance of digestive and mental functions. A dysbiosis of the intestinal microbiota is becoming documented as a factor in the pathogenesis of various pathological conditions, including a plethora of mental, metabolic, and digestive disorders. The diverse etiology of these disorders has been related to different microbes, although insufficient information is currently available on the causal direction of the association. Recent impressive advances in next generation sequencing technologies, along with the progress and innovations of metagenomics, metabolomics, multiomics, bioinformatics, and artificial intelligence tools, have provided prospects to better characterize the microbial populations and their functions and help in better correlation prediction. Moreover, studies using germ-free animals have provided important knowledge on causality rather than association. The research focus needs to be further shifted from individual microbes and their role in influencing health and disease toward the gut microbiome ecosystem. A better understanding of fundamental rules driving interactions within gut microbial communities and the dynamics through which they are acquired, transmitted, and adapted to single individuals is needed to make further progress.
The gut microbiome–brain axis embodies a sophisticated network of biological constructs that scientists are only beginning to understand. The nutritional and therapeutic approaches to modulate this axis are ultimately aimed at improving human quality of life. Some products are even already on the market, including foods and supplements that promise to improve gut, mood, sleep, or cognitive performance. The scientific background behind some of these promises is, however, still arguable or unknown.
The gut microbiome offers interesting possibilities to enhance therapies. Future studies should focus on identifying if gut microbiome signatures correlate with fecal/luminal metabolites and/or cytokines that might translate into marked differences in patients’ life. Moreover, more studies evaluating the potential of therapeutic modulation of the microbiome by biotics/fecal transplant to enhance therapeutic options for diseases are needed. Nowadays, research has shown that fecal transplants can restore healthy bacteria in the lower intestine, which can help control diseases caused by pathogenic bacteria. As a result, the first approved fecal transplant drugs are already a fact. Australia’s Therapeutic Goods Administration was the first to grant approval to biotechnology company BiomeBank for its microbiome-based therapy product Biomictra for treating infections from Clostridioides difficile bacteria. (526,527) Days later, the US FDA also approved its first fecal microbiota product Rebyota produced by Ferring Pharmaceuticals for the prevention of recurrence of C. difficile infection in adults. (528)
The multitude of relations between the microbiota, gut, and brain are now well-documented. The next step─moving on from correlative analysis toward understanding the mechanisms behind these relations and identifying the best ways to adapt and adjust the microbiota for potential therapeutic approaches─is now the required pathway. Some of the key setbacks in existing knowledge include understanding the immunological function of specific microbes in the human gut microbiota and their role in neurodegenerative and psychiatric disorders and how microbial metabolites influence brain function in tandem with immunological and neurological signaling molecules.
Despite the advances in microbiome knowledge, a lack of standardization significantly complicates and obstructs comparisons across studies, thus hampering insights into the structure and function of microbial populations. Recent efforts in the introduction of standardized protocols and analytical methods to characterize microbiota and explore the relationships and co-occurrences of microbially related metabolites and microbial taxa (529) would allow for great improvements in examinations of microbial diversity.
One of the significant challenges in microbiota-based medicine is the delineation of healthy microbiota. Variations in microbiota composition between individuals can be large, i.e., microbiota turn out to be pretty much person-specific, which greatly complicates a “one size fits all” strategy in targeting it. However, it also offers chances since the microbiota might be the outlet for an effective future personalized medicine approach. (530) Overall, clinical studies are hampered by the deficiency of specific biomarkers. Yet, recent meta-analyses have validated a positive assessment of the use of psychobiotic interventions for anxiety, schizophrenia, or cognitive performance, which aim at the diversity and complexity of gut microbiota, as well as the various confounding factors that may affect it. (462,531−536)
Although the prevention of brain disorders still remains out of reach, the knowledge of healthy microbiota and their communication pathways could enable an early prediction of such disorders. Thus, the first signs of neurodegenerative conditions, such as Alzheimer’s and Parkinson’s diseases, are known to develop many years before diagnosis. It might become thinkable to slow down neurodegenerative processes by altering the microbiome. Such possibilities have inspired a growing number of scientists to initiate start-up companies examining therapeutics for the treatment of neurological and other disorders through microbiome modulation. Private investors are showing a strong upward trend to fund such clinical research.
Perfecting the gut microbiota through fecal transplants; pro-, pre-, post-, and syn-biotics; healthy diet; and/or healthy lifestyle to control gut micriobiome–brain axis functions and promote mental and digestive health will be a promising field in the future. Patients suffering from mental and/or digestive disorders will get help through such treatments. Healthy individuals will promote their homeostasis and resilience from these remedies.

Supporting Information

ARTICLE SECTIONS
Jump To

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.3c00127.

  • Prebiotic, probiotic, postbiotic, and fecal transplant therapeutic clinical trial data investigating the treatment of mental disorders and DGBI through gut microbiome modulation (XLSX)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Author
  • Authors
    • Janet M. Sasso - CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesOrcidhttps://orcid.org/0000-0002-1156-5184
    • Ramy M. Ammar - Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
    • Rumiana Tenchov - CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United StatesOrcidhttps://orcid.org/0000-0003-4698-6832
    • Steven Lemmel - CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
    • Olaf Kelber - Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
    • Malte Grieswelle - Bayer Consumer Health, R&D Digestive Health, Darmstadt 64295, Germany
  • Author Contributions

    J.M.S., R.M.A., and R.T. contributed equally to this work.

  • Notes
    The authors declare the following competing financial interest(s): R.M.A and O.K. are employed by Bayer Consumer Health, Germany. M.G. was an intern in Bayer consumer health, Germany during writing this manuscript.

Acknowledgments

ARTICLE SECTIONS
Jump To

The authors sincerely appreciate the CAS Data, Analytics & Insights team for their assistance in data extraction. The authors also appreciate Dharmini Patel for project coordination, Manuela Pausan for ideation, and Peter Jap and Cristina Tomeo for insightful discussion. The authors are also grateful to Manuel Guzman, Gilles Georges, Michael Dennis, Dawn Riedel, Dawn George, and Hong Xie for executive sponsorship.

Abbreviations

ARTICLE SECTIONS
Jump To

4-EPS

4-ethylphenylsulfate

5-AVAB

5-aminovaleric acid betaine

BAs

bile acids

BBB

blood-brain barrier

BCFA

branched-chain fatty acids

CAN

central autonomic network

CEN

central executive network

CNS

central nervous system

DGBI

disorders of gut–brain interaction

DMN

default mode network

EAN

emotional arousal network

ENS

enteric nervous system

FC

functional constipation

FCT

fecal microbiota transplantation

FD

functional dyspepsia

FGIDs

functional gastrointestinal disorders

GBA

gut–brain axis

HC

healthy controls

HPA

hypothalamus–pituitary–adrenal

IBS

irritable bowel syndrome

IHMC

International Human Microbiome Consortium

MPs

muramyl peptides

OCC

occipital network

RDA

recommended daily allowance

SAL

salience network

SCFA

short-chain fatty acids

SMN

sensorimotor network

TMAO

trimethylamine-N-oxide

References

ARTICLE SECTIONS
Jump To

This article references 536 other publications.

  1. 1
    Schopf, J. W. Disparate rates, differing fates: tempo and mode of evolution changed from the Precambrian to the Phanerozoic. Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 67356742,  DOI: 10.1073/pnas.91.15.6735
  2. 2
    Averill, C.; Anthony, M. A.; Baldrian, P.; Finkbeiner, F.; van den Hoogen, J.; Kiers, T.; Kohout, P.; Hirt, E.; Smith, G. R.; Crowther, T. W. Defending Earth’s terrestrial microbiome. Nat. Microbiol. 2022, 7, 1717,  DOI: 10.1038/s41564-022-01228-3
  3. 3
    O’Hara, A. M.; Shanahan, F. The gut flora as a forgotten organ. EMBO reports 2006, 7, 688693,  DOI: 10.1038/sj.embor.7400731
  4. 4
    Bäckhed, F.; Ley, R. E.; Sonnenburg, J. L.; Peterson, D. A.; Gordon, J. I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 19151920,  DOI: 10.1126/science.1104816
  5. 5
    Cénit, M. C.; Matzaraki, V.; Tigchelaar, E. F.; Zhernakova, A. Rapidly expanding knowledge on the role of the gut microbiome in health and disease. Biochim. Biophys. Acta 2014, 1842, 19811992,  DOI: 10.1016/j.bbadis.2014.05.023
  6. 6
    Vrancken, G.; Gregory, A. C.; Huys, G. R. B.; Faust, K.; Raes, J. Synthetic ecology of the human gut microbiota. Nature Reviews Microbiology 2019, 17, 754763,  DOI: 10.1038/s41579-019-0264-8
  7. 7
    Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ. 2019, 7, e7502,  DOI: 10.7717/peerj.7502
  8. 8
    Pariente, N.; York, A. Milestones in human microbiota research. https://www.nature.com/immersive/d42859-019-00041-z/index.html (accessed October 6, 2022).
  9. 9
    Malla, M. A.; Dubey, A.; Kumar, A.; Yadav, S.; Hashem, A.; Abd_Allah, E. F. Exploring the Human Microbiome: The Potential Future Role of Next-Generation Sequencing in Disease Diagnosis and Treatment. Front. Immunol. 2019, 9, 02868,  DOI: 10.3389/fimmu.2018.02868
  10. 10
    Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.-C. C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G. H. Microbiome definition re-visited: old concepts and new challenges. Microbiome 2020, 8, 103,  DOI: 10.1186/s40168-020-00875-0
  11. 11
    Butler, M. I.; Mörkl, S.; Sandhu, K. V.; Cryan, J. F.; Dinan, T. G. The Gut Microbiome and Mental Health: What Should We Tell Our Patients?: Le microbiote Intestinal et la Santé Mentale: que Devrions-Nous dire à nos Patients?. Can. J. Psychiatry. 2019, 64, 747760,  DOI: 10.1177/0706743719874168
  12. 12
    Corliss, J. O. Three Centuries of Protozoology: A Brief Tribute to its Founding Father, A. van Leeuwenhoek of Delft. Journal of Protozoology 1975, 22, 37,  DOI: 10.1111/j.1550-7408.1975.tb00934.x
  13. 13
    Leidy, J. A flora and fauna within living animals. Smithsonian Institution: WA, 1853.
  14. 14
    Savage, D. C. Microbial biota of the human intestine: a tribute to some pioneering scientists. Curr. Issues Intest. Microbiol. 2001, 2, 115
  15. 15
    Pariente, N. A field is born. 2019. https://www.nature.com/articles/d42859-019-00006-2.
  16. 16
    Micheli, P. A. Nova plantarum genera iuxta Tournefortii methodum disposita ; 1729.
  17. 17
    Drews, G. The roots of microbiology and the influence of Ferdinand Cohn on microbiology of the 19th century. FEMS Microbiol. Rev. 2000, 24, 225249,  DOI: 10.1111/j.1574-6976.2000.tb00540.x
  18. 19
    Metchnikoff, E. Prolongation of Life: Optimistic Studies ; 1923.
  19. 20
    Sonnenborn, U. Escherichia coli strain Nissle 1917─from bench to bedside and back: history of a special Escherichia coli strain with probiotic properties. FEMS Microbiol. Lett. 2016, 363, fnw212,  DOI: 10.1093/femsle/fnw212
  20. 21
    Tan, S. Y.; Tatsumura, Y. Alexander Fleming (1881–1955): Discoverer of penicillin. Singapore Med. J. 2015, 56, 366367,  DOI: 10.11622/smedj.2015105
  21. 22
    Almeida, A.; Mitchell, A. L.; Boland, M.; Forster, S. C.; Gloor, G. B.; Tarkowska, A.; Lawley, T. D.; Finn, R. D. A new genomic blueprint of the human gut microbiota. Nature 2019, 568, 499504,  DOI: 10.1038/s41586-019-0965-1
  22. 23
    Hungate, R. E., Chapter IV A Roll Tube Method for Cultivation of Strict Anaerobes. In Methods in Microbiology, Vol. 3; Norris, J. R., Ribbons, D. W., Eds.; Academic Press, 1969; pp 117132.
  23. 24
    Cat, L. A. The Decade Of The Microbiome. https://www.forbes.com/sites/linhanhcat/2019/12/31/decade-of-the-microbiome/?sh=7e621a679961 (accessed October 11, 2022).
  24. 25
    NIH Human Microbiome Project. https://www.hmpdacc.org/overview/ (accessed October 6, 2022).
  25. 26
    MetaHIT. https://www.gutmicrobiotaforhealth.com/metahit/ (accessed October 6, 2022).
  26. 27
    Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K. S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 5965,  DOI: 10.1038/nature08821
  27. 28
    Proctor, Lita M. The Human Microbiome Project in 2011 and Beyond. Cell Host Microbe 2011, 10, 287291,  DOI: 10.1016/j.chom.2011.10.001
  28. 29
    Birney, E.; Stamatoyannopoulos, J. A.; Dutta, A.; Guigó, R.; Gingeras, T. R.; Margulies, E. H.; Weng, Z.; Snyder, M.; Dermitzakis, E. T.; Stamatoyannopoulos, J. A. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799816,  DOI: 10.1038/nature05874
  29. 30
    Riesenfeld, C. S.; Schloss, P. D.; Handelsman, J. Metagenomics: Genomic Analysis of Microbial Communities. Annu. Rev. Genet. 2004, 38, 525552,  DOI: 10.1146/annurev.genet.38.072902.091216
  30. 31
    Jandhyala, S. M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 87878803,  DOI: 10.3748/wjg.v21.i29.8787
  31. 32
    Tomas, J.; Wrzosek, L.; Bouznad, N.; Bouet, S.; Mayeur, C.; Noordine, M. L.; Honvo-Houeto, E.; Langella, P.; Thomas, M.; Cherbuy, C. Primocolonization is associated with colonic epithelial maturation during conventionalization. FASEB J. 2013, 27, 645655,  DOI: 10.1096/fj.12-216861
  32. 33
    Caballero, S.; Pamer, E. G. Microbiota-mediated inflammation and antimicrobial defense in the intestine. Annu. Rev. Immunol. 2015, 33, 227256,  DOI: 10.1146/annurev-immunol-032713-120238
  33. 34
    Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Frontiers in Genetics 2015, 6, 148,  DOI: 10.3389/fgene.2015.00148
  34. 35
    Mayer, E. A.; Knight, R.; Mazmanian, S. K.; Cryan, J. F.; Tillisch, K. Gut microbes and the brain: paradigm shift in neuroscience. J. Neurosci. 2014, 34, 1549015496,  DOI: 10.1523/JNEUROSCI.3299-14.2014
  35. 36
    Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R. D.; Shanahan, F.; Dinan, T. G.; Cryan, J. F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666673,  DOI: 10.1038/mp.2012.77
  36. 37
    Bray, N. The microbiota–gut–brain axis. https://www.nature.com/articles/d42859-019-00021-3 (accessed October 6, 2022).
  37. 38
    Dinan, T. G.; Cryan, J. F. The impact of gut microbiota on brain and behaviour: implications for psychiatry. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 552558,  DOI: 10.1097/MCO.0000000000000221
  38. 39
    Bravo, J. A.; Forsythe, P.; Chew, M. V.; Escaravage, E.; Savignac, H. M.; Dinan, T. G.; Bienenstock, J.; Cryan, J. F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 1605016055,  DOI: 10.1073/pnas.1102999108
  39. 40
    Cohen-Sacks, H.; Elazar, V.; Gao, J.; Golomb, A.; Adwan, H.; Korchov, N.; Levy, R. J.; Berger, M. R.; Golomb, G. Delivery and expression of pDNA embedded in collagen matrices. J. Controlled Release 2004, 95, 309320,  DOI: 10.1016/j.jconrel.2003.11.001
  40. 41
    Rutsch, A.; Kantsjö, J. B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179,  DOI: 10.3389/fimmu.2020.604179
  41. 42
    Drossman, D. A.; Hasler, W. L. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. Gastroenterology 2016, 150, 12571261,  DOI: 10.1053/j.gastro.2016.03.035
  42. 43
    Perez-Muñoz, M. E.; Arrieta, M.-C.; Ramer-Tait, A. E.; Walter, J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017, 5, 48,  DOI: 10.1186/s40168-017-0268-4
  43. 44
    Dominguez-Bello, M. G.; Costello, E. K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 1197111975,  DOI: 10.1073/pnas.1002601107
  44. 45
    Hill, C. J.; Lynch, D. B.; Murphy, K.; Ulaszewska, M.; Jeffery, I. B.; O’Shea, C. A.; Watkins, C.; Dempsey, E.; Mattivi, F.; Tuohy, K. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome 2017, 5, 4,  DOI: 10.1186/s40168-016-0213-y
  45. 46
    Vatanen, T.; Jabbar, K. S.; Ruohtula, T.; Honkanen, J.; Avila-Pacheco, J.; Siljander, H.; Stražar, M.; Oikarinen, S.; Hyöty, H.; Ilonen, J. Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolism. Cell 2022, 185, 49214936.e4915,  DOI: 10.1016/j.cell.2022.11.023
  46. 47
    Baleato, C. L.; Ferguson, J. J. A.; Oldmeadow, C.; Mishra, G. D.; Garg, M. L. Plant-Based Dietary Patterns versus Meat Consumption and Prevalence of Impaired Glucose Intolerance and Diabetes Mellitus: A Cross-Sectional Study in Australian Women. Nutrients 2022, 14, 4152,  DOI: 10.3390/nu14194152
  47. 48
    Keaver, L.; Ruan, M.; Chen, F.; Du, M.; Ding, C.; Wang, J.; Shan, Z.; Liu, J.; Zhang, F. F. Plant- and animal-based diet quality and mortality among US adults: a cohort study. Br. J. Nutr. 2021, 125, 14051415,  DOI: 10.1017/S0007114520003670
  48. 49
    O’Mahony, S. M.; Marchesi, J. R.; Scully, P.; Codling, C.; Ceolho, A.-M.; Quigley, E. M. M.; Cryan, J. F.; Dinan, T. G. Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses. Biol. Psychiatry 2009, 65, 263267,  DOI: 10.1016/j.biopsych.2008.06.026
  49. 50
    Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J. K. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology (Reading) 2010, 156, 32163223,  DOI: 10.1099/mic.0.040618-0
  50. 51
    Caporaso, J. G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F. D.; Costello, E. K.; Fierer, N.; Peña, A. G.; Goodrich, J. K.; Gordon, J. I. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 2010, 7, 335336,  DOI: 10.1038/nmeth.f.303
  51. 52
    Tang, L. Sequence-based identification of human-associated microbiota. https://www.nature.com/articles/d42859-019-00011-5 (accessed October 24, 2022).
  52. 53
    Collins, S. M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735742,  DOI: 10.1038/nrmicro2876
  53. 54
    Lagier, J.-C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M. Microbial culturomics: paradigm shift in the human gut microbiome study. Clinical Microbiology and Infection 2012, 18, 11851193,  DOI: 10.1111/1469-0691.12023
  54. 55
    Smith, P. M.; Howitt, M. R.; Panikov, N.; Michaud, M.; Gallini, C. A.; Bohlooly, Y. M.; Glickman, J. N.; Garrett, W. S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569573,  DOI: 10.1126/science.1241165
  55. 56
    Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232236,  DOI: 10.1038/nature12331
  56. 57
    Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J. R.; Pfeffer, K.; Coffer, P. J. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451455,  DOI: 10.1038/nature12726
  57. 58
    Donia, M. S.; Cimermancic, P.; Schulze, C. J.; Wieland Brown, L. C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R. G.; Fischbach, M. A. A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibiotics. Cell 2014, 158, 14021414,  DOI: 10.1016/j.cell.2014.08.032
  58. 59
    Lurie, I.; Yang, Y. X.; Haynes, K.; Mamtani, R.; Boursi, B. Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J. Clin. Psychiatry 2015, 76, 15221528,  DOI: 10.4088/JCP.15m09961
  59. 60
    Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786796,  DOI: 10.1038/mp.2016.44
  60. 61
    Davis, D. J.; Hecht, P. M.; Jasarevic, E.; Beversdorf, D. Q.; Will, M. J.; Fritsche, K.; Gillespie, C. H. Sex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated mice. Brain, Behavior, and Immunity 2017, 59, 3848,  DOI: 10.1016/j.bbi.2016.09.003
  61. 62
    Pasolli, E.; Asnicar, F.; Manara, S.; Zolfo, M.; Karcher, N.; Armanini, F.; Beghini, F.; Manghi, P.; Tett, A.; Ghensi, P. Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and Lifestyle. Cell 2019, 176, 649662.e20,  DOI: 10.1016/j.cell.2019.01.001
  62. 63
    Nayfach, S.; Shi, Z. J.; Seshadri, R.; Pollard, K. S.; Kyrpides, N. C. New insights from uncultivated genomes of the global human gut microbiome. Nature 2019, 568, 505510,  DOI: 10.1038/s41586-019-1058-x
  63. 64
    Pusceddu, M. M.; Barboza, M.; Schneider, M.; Stokes, P.; Sladek, J. A.; Torres-Fuentes, C.; Goldfild, L. R.; Gillis, S. E.; Brust-Mascher, I.; Rabasa, G. Nod-like receptors are critical for gut-brain axis signaling in mice. J. Physiol. 2019, 597, 57775797,  DOI: 10.1113/JP278640
  64. 65
    Pellegrini, C.; Antonioli, L.; Calderone, V.; Colucci, R.; Fornai, M.; Blandizzi, C. Microbiota-gut-brain axis in health and disease: Is NLRP3 inflammasome at the crossroads of microbiota-gut-brain communications?. Prog. Neurobiol. 2020, 191, 101806,  DOI: 10.1016/j.pneurobio.2020.101806
  65. 66
    Zhao, K.; Yao, M.; Zhang, X.; Xu, F.; Shao, X.; Wei, Y.; Wang, H. Flavonoids and intestinal microbes interact to alleviate depression. Journal of the Science of Food and Agriculture 2022, 102, 13111318,  DOI: 10.1002/jsfa.11578
  66. 67
    Segre, J. A. What does it take to satisfy Koch’s postulates two centuries later? Microbial genomics and Propionibacteria acnes. J. Invest. Dermatol. 2013, 133, 21412142,  DOI: 10.1038/jid.2013.260
  67. 68
    Eiseman, B.; Silen, W.; Bascom, G. S.; Kauvar, A. J. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958, 44, 854859
  68. 69
    Reyniers, J. A. The pure-culture concept and gnotobiotics. Ann. N.Y. Acad. Sci. 1959, 78, 316,  DOI: 10.1111/j.1749-6632.1959.tb53091.x
  69. 70
    Schaedler, R. W.; Dubos, R.; Costello, R. ASSOCIATION OF GERMFREE MICE WITH BACTERIA ISOLATED FROM NORMAL MICE. J. Exp. Med. 1965, 122, 7782,  DOI: 10.1084/jem.122.1.77
  70. 71
    Peppercorn, M. A.; Goldman, P. The Role of Intestinal Bacteria in the Metabolism of Salicylazosulfapyridine. Journal of Pharmacology and Experimental Therapeutics 1972, 181, 555562
  71. 72
    Stark, P. L.; Lee, A. The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J. Med. Microbiol. 1982, 15, 189203,  DOI: 10.1099/00222615-15-2-189
  72. 73
    Fleischmann, R. D.; Adams, M. D.; White, O.; Clayton, R. A.; Kirkness, E. F.; Kerlavage, A. R.; Bult, C. J.; Tomb, J. F.; Dougherty, B. A.; Merrick, J. M. Whole-genome random sequencing and assembly of Haemophilus influenzae Rd. Science 1995, 269, 496512,  DOI: 10.1126/science.7542800
  73. 74
    Wilson, K. H.; Blitchington, R. B. Human colonic biota studied by ribosomal DNA sequence analysis. Appl. Environ. Microbiol. 1996, 62, 22732278,  DOI: 10.1128/aem.62.7.2273-2278.1996
  74. 75
    Guarner, F.; Malagelada, J. R. Gut flora in health and disease. Lancet 2003, 361, 512519,  DOI: 10.1016/S0140-6736(03)12489-0
  75. 76
    Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229241,  DOI: 10.1016/j.cell.2004.07.002
  76. 77
    Mazmanian, S. K.; Liu, C. H.; Tzianabos, A. O.; Kasper, D. L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107118,  DOI: 10.1016/j.cell.2005.05.007
  77. 78
    Turnbaugh, P. J.; Ley, R. E.; Mahowald, M. A.; Magrini, V.; Mardis, E. R.; Gordon, J. I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 10271031,  DOI: 10.1038/nature05414
  78. 79
    Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C. P. M.; Alou, M. T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M. P. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 9197,  DOI: 10.1126/science.aan3706
  79. 80
    Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M. L.; Luke, J. J.; Gajewski, T. F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104108,  DOI: 10.1126/science.aao3290
  80. 81
    Belizário, J. E.; Napolitano, M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front. Microbiol. 2015, 6, 1050,  DOI: 10.3389/fmicb.2015.01050
  81. 82
    Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G. A. D.; Gasbarrini, A.; Mele, M. C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14,  DOI: 10.3390/microorganisms7010014
  82. 83
    Cresci, G. A. M.; Izzo, K. Chapter 4 - Gut Microbiome. In Adult Short Bowel Syndrome; Corrigan, M. L., Roberts, K., Steiger, E., Eds.; Academic Press, 2019; pp 4554.
  83. 84
    CAS Content Collection. https://www.cas.org/about/cas-content (accessed January 18, 2023).
  84. 85
    Kho, Z. Y.; Lal, S. K. The Human Gut Microbiome – A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835,  DOI: 10.3389/fmicb.2018.01835
  85. 86
    Sankar, S. A.; Lagier, J. C.; Pontarotti, P.; Raoult, D.; Fournier, P. E. The human gut microbiome, a taxonomic conundrum. Syst. Appl. Microbiol 2015, 38, 276286,  DOI: 10.1016/j.syapm.2015.03.004
  86. 87
    Manson, J. M.; Rauch, M.; Gilmore, M. S. The Commensal Microbiology of the Gastrointestinal Tract. In GI Microbiota and Regulation of the Immune System; Huffnagle, G. B., Noverr, M. C., Eds.; Landes Bioscience and Springer Science+Business Media, LLC: New York, 2008; pp 1528.
  87. 88
    Kim, D.-H. Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metab. Dispos. 2015, 43, 15811589,  DOI: 10.1124/dmd.115.063867
  88. 89
    Deng, P.; Swanson, K. S. Gut microbiota of humans, dogs and cats: current knowledge and future opportunities and challenges. Br. J. Nutr. 2015, 113 (Suppl), S617,  DOI: 10.1017/S0007114514002943
  89. 90
    Nardone, G.; Compare, D. The human gastric microbiota: Is it time to rethink the pathogenesis of stomach diseases?. United European Gastroenterol J. 2015, 3, 255260,  DOI: 10.1177/2050640614566846
  90. 91
    Ghosh, S.; Pramanik, S. Structural diversity, functional aspects and future therapeutic applications of human gut microbiome. Arch. Microbiol. 2021, 203, 52815308,  DOI: 10.1007/s00203-021-02516-y
  91. 92
    Leser, T. D.; Mo̷lbak, L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ. Microbiol. 2009, 11, 21942206,  DOI: 10.1111/j.1462-2920.2009.01941.x
  92. 93
    Linares, D. M.; Ross, P.; Stanton, C. Beneficial Microbes: The pharmacy in the gut. Bioengineered 2016, 7, 1120,  DOI: 10.1080/21655979.2015.1126015
  93. 94
    Shah, H. N.; Olsen, I.; Bernard, K.; Finegold, S. M.; Gharbia, S.; Gupta, R. S. Approaches to the study of the systematics of anaerobic, Gram-negative, non-sporeforming rods: Current status and perspectives. Anaerobe 2009, 15, 179194,  DOI: 10.1016/j.anaerobe.2009.08.003
  94. 95
    Thomas, F.; Hehemann, J.-H.; Rebuffet, E.; Czjzek, M.; Michel, G. Environmental and Gut Bacteroidetes: The Food Connection. Front. Microbiol. 2011, 2, 93,  DOI: 10.3389/fmicb.2011.00093
  95. 96
    McKee, L. S.; La Rosa, S. L.; Westereng, B.; Eijsink, V. G.; Pope, P. B.; Larsbrink, J. Polysaccharide degradation by the Bacteroidetes: mechanisms and nomenclature. Environ. Microbiol. Rep. 2021, 13, 559581,  DOI: 10.1111/1758-2229.12980
  96. 97
    Ludwig, W.; Schleifer, K.-H.; Whitman, W. B. Revised road map to the phylum Firmicutes. In Bergey’s Manual of Systematic Bacteriology: Vol. Three The Firmicutes; De Vos, P., Garrity, G. M., Jones, D., Krieg, N. R., Ludwig, W., Rainey, F. A., Schleifer, K.-H., Whitman, W. B., Eds.; Springer New York: New York, 2009; pp 113.
  97. 98
    Yang, M.; Gu, Y.; Li, L.; Liu, T.; Song, X.; Sun, Y.; Cao, X.; Wang, B.; Jiang, K.; Cao, H. Bile Acid–Gut Microbiota Axis in Inflammatory Bowel Disease: From Bench to Bedside. Nutrients 2021, 13, 3143,  DOI: 10.3390/nu13093143
  98. 99
    Barka, E. A.; Vatsa, P.; Sanchez, L.; Gaveau-Vaillant, N.; Jacquard, C.; Klenk, H.-P.; Clément, C.; Ouhdouch, Y.; Wezel, G. P. v. Taxonomy, Physiology, and Natural Products of Actinobacteria. Microbiology and Molecular Biology Reviews 2016, 80, 143,  DOI: 10.1128/MMBR.00019-15
  99. 100
    Hidalgo-Cantabrana, C.; Delgado, S.; Ruiz, L.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A. Bifidobacteria and Their Health-Promoting Effects. Microbiol Spectr 2017, 5, 5.3.21,  DOI: 10.1128/microbiolspec.BAD-0010-2016
  100. 101
    Stackebrandt, E.; Murray, R. G. E.; Trüper, H. G. Proteobacteria classis nov., a Name for the Phylogenetic Taxon That Includes the “Purple Bacteria and Their Relatives. International Journal of Systematic and Evolutionary Microbiology 1988, 38, 321325,  DOI: 10.1099/00207713-38-3-321
  101. 102
    Rizzatti, G.; Lopetuso, L. R.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A Common Factor in Human Diseases. BioMed. Research International 2017, 2017, 9351507,  DOI: 10.1155/2017/9351507
  102. 103
    Lee, K.-C.; Webb, R. I.; Janssen, P. H.; Sangwan, P.; Romeo, T.; Staley, J. T.; Fuerst, J. A. Phylum Verrucomicrobia representatives share a compartmentalized cell plan with members of bacterial phylum Planctomycetes. BMC Microbiol. 2009, 9, 5,  DOI: 10.1186/1471-2180-9-5
  103. 104
    Rappé, M. S.; Giovannoni, S. J. The Uncultured Microbial Majority. Annu. Rev. Microbiol. 2003, 57, 369394,  DOI: 10.1146/annurev.micro.57.030502.090759
  104. 105
    Geerlings, S. Y.; Ouwerkerk, J. P.; Koehorst, J. J.; Ritari, J.; Aalvink, S.; Stecher, B.; Schaap, P. J.; Paulin, L.; de Vos, W. M.; Belzer, C. Genomic convergence between Akkermansia muciniphila in different mammalian hosts. BMC Microbiol. 2021, 21, 298,  DOI: 10.1186/s12866-021-02360-6
  105. 106
    Liu, M.-J.; Yang, J.-Y.; Yan, Z.-H.; Hu, S.; Li, J.-Q.; Xu, Z.-X.; Jian, Y.-P. Recent findings in Akkermansia muciniphila-regulated metabolism and its role in intestinal diseases. Clin. Nutr. 2022, 41, 23332344,  DOI: 10.1016/j.clnu.2022.08.029
  106. 107
    Gupta, R. S.; Sethi, M. Phylogeny and molecular signatures for the phylum Fusobacteria and its distinct subclades. Anaerobe 2014, 28, 182198,  DOI: 10.1016/j.anaerobe.2014.06.007
  107. 108
    Madhogaria, B.; Bhowmik, P.; Kundu, A. Correlation between human gut microbiome and diseases. Infectious Medicine 2022, 1, 180191,  DOI: 10.1016/j.imj.2022.08.004
  108. 109
    Bull, M. J.; Plummer, N. T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. 2014, 13, 1722
  109. 110
    Vijay, A.; Valdes, A. M. Role of the gut microbiome in chronic diseases: a narrative review. Eur. J. Clin. Nutr. 2022, 76, 489501,  DOI: 10.1038/s41430-021-00991-6
  110. 111
    Baquero, F.; Nombela, C. The microbiome as a human organ. Clin Microbiol Infect 2012, 18 (Suppl 4), 24,  DOI: 10.1111/j.1469-0691.2012.03916.x
  111. 112
    Evans, J. M.; Morris, L. S.; Marchesi, J. R. The gut microbiome: the role of a virtual organ in the endocrinology of the host. J. Endocrinol. 2013, 218, R37R47,  DOI: 10.1530/JOE-13-0131
  112. 113
    Haseeb, A.; Shahzad, I.; Ghulam, H.; Muhammad Naeem, F.; Humaira, M.; Imtiaz, M.; Imran, M.; Saima, M.; Muhammad Irfan, U. Gut Microbiome: A New Organ System in Body. In Parasitology and Microbiology Research; Gilberto Antonio Bastidas, P., Asghar Ali, K., Eds.; IntechOpen: Rijeka, Croatia, 2019.
  113. 114
    Cani, P. D. Human gut microbiome: hopes, threats and promises. Gut 2018, 67, 17161725,  DOI: 10.1136/gutjnl-2018-316723
  114. 115
    Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A. N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T. W.; Andrews, E.; Ajami, N. J.; Bonham, K. S.; Brislawn, C. J. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655662,  DOI: 10.1038/s41586-019-1237-9
  115. 116
    Mars, R. A. T.; Yang, Y.; Ward, T.; Houtti, M.; Priya, S.; Lekatz, H. R.; Tang, X.; Sun, Z.; Kalari, K. R.; Korem, T. Longitudinal Multi-omics Reveals Subset-Specific Mechanisms Underlying Irritable Bowel Syndrome. Cell 2020, 182, 14601473.e17,  DOI: 10.1016/j.cell.2020.08.007
  116. 117
    Rinttilä, T.; Lyra, A.; Krogius-Kurikka, L.; Palva, A. Real-time PCR analysis of enteric pathogens from fecal samples of irritable bowel syndrome subjects. Gut Pathog. 2011, 3, 6,  DOI: 10.1186/1757-4749-3-6
  117. 118
    Mayer, E. A.; Nance, K.; Chen, S. The Gut-Brain Axis. Annu. Rev. Med. 2022, 73, 439453,  DOI: 10.1146/annurev-med-042320-014032
  118. 119
    Zhang, T.; Zhang, C.; Zhang, J.; Sun, F.; Duan, L. Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Front Cell Infect Microbiol 2022, 12, 859967,  DOI: 10.3389/fcimb.2022.859967
  119. 120
    Ponnusamy, K.; Choi, J. N.; Kim, J.; Lee, S. Y.; Lee, C. H. Microbial community and metabolomic comparison of irritable bowel syndrome faeces. J. Med. Microbiol. 2011, 60, 817827,  DOI: 10.1099/jmm.0.028126-0
  120. 121
    Rajilić-Stojanović, M.; Biagi, E.; Heilig, H. G.; Kajander, K.; Kekkonen, R. A.; Tims, S.; de Vos, W. M. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011, 141, 17921801,  DOI: 10.1053/j.gastro.2011.07.043
  121. 122
    Pittayanon, R.; Lau, J. T.; Yuan, Y.; Leontiadis, G. I.; Tse, F.; Surette, M.; Moayyedi, P. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. Gastroenterology 2019, 157, 97108,  DOI: 10.1053/j.gastro.2019.03.049
  122. 123
    El-Salhy, M.; Hatlebakk, J. G.; Gilja, O. H.; Bråthen Kristoffersen, A.; Hausken, T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 2020, 69, 859867,  DOI: 10.1136/gutjnl-2019-319630
  123. 124
    Wilson, B.; Rossi, M.; Kanno, T.; Parkes, G. C.; Anderson, S.; Mason, A. J.; Irving, P. M.; Lomer, M. C.; Whelan, K. β-Galactooligosaccharide in Conjunction With Low FODMAP Diet Improves Irritable Bowel Syndrome Symptoms but Reduces Fecal Bifidobacteria. Am. J. Gastroenterol. 2020, 115, 906915,  DOI: 10.14309/ajg.0000000000000641
  124. 125
    Vasant, D. H.; Paine, P. A.; Black, C. J.; Houghton, L. A.; Everitt, H. A.; Corsetti, M.; Agrawal, A.; Aziz, I.; Farmer, A. D.; Eugenicos, M. P. British Society of Gastroenterology guidelines on the management of irritable bowel syndrome. Gut 2021, 70, 12141240,  DOI: 10.1136/gutjnl-2021-324598
  125. 126
    Moayyedi, P.; Andrews, C. N.; MacQueen, G.; Korownyk, C.; Marsiglio, M.; Graff, L.; Kvern, B.; Lazarescu, A.; Liu, L.; Paterson, W. G. Canadian Association of Gastroenterology Clinical Practice Guideline for the Management of Irritable Bowel Syndrome (IBS). J. Can. Assoc Gastroenterol 2019, 2, 629,  DOI: 10.1093/jcag/gwy071
  126. 127
    Fukudo, S.; Okumura, T.; Inamori, M.; Okuyama, Y.; Kanazawa, M.; Kamiya, T.; Sato, K.; Shiotani, A.; Naito, Y.; Fujikawa, Y. Evidence-based clinical practice guidelines for irritable bowel syndrome 2020. J. Gastroenterol. 2021, 56, 193217,  DOI: 10.1007/s00535-020-01746-z
  127. 128
    Lacy, B. E.; Pimentel, M.; Brenner, D. M.; Chey, W. D.; Keefer, L. A.; Long, M. D.; Moshiree, B. ACG Clinical Guideline: Management of Irritable Bowel Syndrome. Am. J. Gastroenterol. 2021, 116, 17,  DOI: 10.14309/ajg.0000000000001036
  128. 129
    Ford, A. C.; Harris, L. A.; Lacy, B. E.; Quigley, E. M. M.; Moayyedi, P. Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2018, 48, 10441060,  DOI: 10.1111/apt.15001
  129. 130
    Caruso, R.; Lo, B. C.; Núñez, G. Host-microbiota interactions in inflammatory bowel disease. Nat. Rev. Immunol. 2020, 20, 411426,  DOI: 10.1038/s41577-019-0268-7
  130. 131
    Federici, S.; Kredo-Russo, S.; Valdés-Mas, R.; Kviatcovsky, D.; Weinstock, E.; Matiuhin, Y.; Silberberg, Y.; Atarashi, K.; Furuichi, M.; Oka, A. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell 2022, 185, 28792898.e24,  DOI: 10.1016/j.cell.2022.07.003
  131. 132
    Nagalingam, N. A.; Lynch, S. V. Role of the microbiota in inflammatory bowel diseases. Inflamm. Bowel Dis. 2012, 18, 968984,  DOI: 10.1002/ibd.21866
  132. 133
    Barcenilla, A.; Pryde, S. E.; Martin, J. C.; Duncan, S. H.; Stewart, C. S.; Henderson, C.; Flint, H. J. Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl. Environ. Microbiol. 2000, 66, 16541661,  DOI: 10.1128/AEM.66.4.1654-1661.2000
  133. 134
    Halfvarson, J.; Brislawn, C. J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W. A.; Bramer, L. M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol 2017, 2, 17004,  DOI: 10.1038/nmicrobiol.2017.4
  134. 135
    Mottawea, W.; Chiang, C.-K.; Mühlbauer, M.; Starr, A. E.; Butcher, J.; Abujamel, T.; Deeke, S. A.; Brandel, A.; Zhou, H.; Shokralla, S. Altered intestinal microbiota–host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 2016, 7, 13419,  DOI: 10.1038/ncomms13419
  135. 136
    Marchesi, J. R.; Holmes, E.; Khan, F.; Kochhar, S.; Scanlan, P.; Shanahan, F.; Wilson, I. D.; Wang, Y. Rapid and noninvasive metabonomic characterization of inflammatory bowel disease. J. Proteome Res. 2007, 6, 546551,  DOI: 10.1021/pr060470d
  136. 137
    Van Immerseel, F.; Ducatelle, R.; De Vos, M.; Boon, N.; Van De Wiele, T.; Verbeke, K.; Rutgeerts, P.; Sas, B.; Louis, P.; Flint, H. J. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. J. Med. Microbiol. 2010, 59, 141143,  DOI: 10.1099/jmm.0.017541-0
  137. 138
    Miquel, S.; Martín, R.; Rossi, O.; Bermúdez-Humarán, L. G.; Chatel, J. M.; Sokol, H.; Thomas, M.; Wells, J. M.; Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013, 16, 255261,  DOI: 10.1016/j.mib.2013.06.003
  138. 139
    Tamanai-Shacoori, Z.; Smida, I.; Bousarghin, L.; Loreal, O.; Meuric, V.; Fong, S. B.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Roseburia spp.: a marker of health?. Future Microbiol. 2017, 12, 157170,  DOI: 10.2217/fmb-2016-0130
  139. 140
    Li, Y.; Xia, S.; Jiang, X.; Feng, C.; Gong, S.; Ma, J.; Fang, Z.; Yin, J.; Yin, Y. Gut Microbiota and Diarrhea: An Updated Review. Microbiology 2021, 11, 625210,  DOI: 10.3389/fcimb.2021.625210
  140. 141
    Hodges, K.; Gill, R. Infectious diarrhea: Cellular and molecular mechanisms. Gut Microbes 2010, 1, 421,  DOI: 10.4161/gmic.1.1.11036
  141. 142
    Levine, M. M. Escherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherent. J. Infect. Dis. 1987, 155, 377389,  DOI: 10.1093/infdis/155.3.377
  142. 143
    Gallo, A.; Passaro, G.; Gasbarrini, A.; Landolfi, R.; Montalto, M. Modulation of microbiota as treatment for intestinal inflammatory disorders: An uptodate. World J. Gastroenterol. 2016, 22, 71867202,  DOI: 10.3748/wjg.v22.i32.7186
  143. 144
    Bron, P. A.; Kleerebezem, M.; Brummer, R. J.; Cani, P. D.; Mercenier, A.; MacDonald, T. T.; Garcia-Ródenas, C. L.; Wells, J. M. Can probiotics modulate human disease by impacting intestinal barrier function?. Br. J. Nutr. 2017, 117, 93107,  DOI: 10.1017/S0007114516004037
  144. 145
    Zhang, S.; Wang, R.; Li, D.; Zhao, L.; Zhu, L. Role of gut microbiota in functional constipation. Gastroenterol. Rep. 2021, 9, 392401,  DOI: 10.1093/gastro/goab035
  145. 146
    Zoppi, G.; Cinquetti, M.; Luciano, A.; Benini, A.; Muner, A.; Bertazzoni Minelli, E. The intestinal ecosystem in chronic functional constipation. Acta Paediatr. 1998, 87, 836841,  DOI: 10.1111/j.1651-2227.1998.tb01547.x
  146. 147
    Khalif, I. L.; Quigley, E. M.; Konovitch, E. A.; Maximova, I. D. Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation. Dig. Liver Dis. 2005, 37, 838849,  DOI: 10.1016/j.dld.2005.06.008
  147. 148
    Kim, S. E.; Choi, S. C.; Park, K. S.; Park, M. I.; Shin, J. E.; Lee, T. H.; Jung, K. W.; Koo, H. S.; Myung, S. J. Change of Fecal Flora and Effectiveness of the Short-term VSL#3 Probiotic Treatment in Patients With Functional Constipation. J. Neurogastroenterol. Motil. 2015, 21, 111120,  DOI: 10.5056/jnm14048
  148. 149
    Zhu, L.; Liu, W.; Alkhouri, R.; Baker, R. D.; Bard, J. E.; Quigley, E. M.; Baker, S. S. Structural changes in the gut microbiome of constipated patients. Physiol. Genomics 2014, 46, 679686,  DOI: 10.1152/physiolgenomics.00082.2014
  149. 150
    Mancabelli, L.; Milani, C.; Lugli, G. A.; Turroni, F.; Mangifesta, M.; Viappiani, A.; Ticinesi, A.; Nouvenne, A.; Meschi, T.; van Sinderen, D. Unveiling the gut microbiota composition and functionality associated with constipation through metagenomic analyses. Sci. Rep. 2017, 7, 9879,  DOI: 10.1038/s41598-017-10663-w
  150. 151
    Naseer, M.; Poola, S.; Uraz, S.; Tahan, V. Therapeutic Effects of Prebiotics on Constipation: A Schematic Review. Curr. Clin. Pharmacol. 2020, 15, 207215,  DOI: 10.2174/1574884715666200212125035
  151. 152
    Zhao, Y.; Yu, Y. B. Intestinal microbiota and chronic constipation. SpringerPlus 2016, 5, 1130,  DOI: 10.1186/s40064-016-2821-1
  152. 153
    Wang, J. K.; Yao, S. K. Roles of Gut Microbiota and Metabolites in Pathogenesis of Functional Constipation. Evid. Based Complement. Alternat. Med. 2021, 2021, 5560310,  DOI: 10.1155/2021/5560310
  153. 154
    Rao, S. S.; Rattanakovit, K.; Patcharatrakul, T. Diagnosis and management of chronic constipation in adults. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 295305,  DOI: 10.1038/nrgastro.2016.53
  154. 155
    Moloney, R. D.; Desbonnet, L.; Clarke, G.; Dinan, T. G.; Cryan, J. F. The microbiome: stress, health and disease. Mamm. Genome 2014, 25, 4974,  DOI: 10.1007/s00335-013-9488-5
  155. 156
    Mayer, E. A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Invest. 2015, 125, 926938,  DOI: 10.1172/JCI76304
  156. 157
    Maldonado-Contreras, A.; Noel, S. E.; Ward, D. V.; Velez, M.; Mangano, K. M. Associations between Diet, the Gut Microbiome, and Short-Chain Fatty Acid Production among Older Caribbean Latino Adults. J. Acad. Nutr. Diet. 2020, 120, 20472060.e46,  DOI: 10.1016/j.jand.2020.04.018
  157. 158
    Zhang, H.; Chen, Y.; Wang, Z.; Xie, G.; Liu, M.; Yuan, B.; Chai, H.; Wang, W.; Cheng, P. Implications of Gut Microbiota in Neurodegenerative Diseases. Front. Immunol. 2022, 13, 785644,  DOI: 10.3389/fimmu.2022.785644
  158. 159
    Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98104,  DOI: 10.1016/j.jneuroim.2019.01.004
  159. 160
    Caspani, G.; Kennedy, S.; Foster, J. A.; Swann, J. Gut microbial metabolites in depression: understanding the biochemical mechanisms. Microbial cell (Graz, Austria) 2019, 6, 454481,  DOI: 10.15698/mic2019.10.693
  160. 161
    Roubalová, R.; Procházková, P.; Papežová, H.; Smitka, K.; Bilej, M.; Tlaskalová-Hogenová, H. Anorexia nervosa: Gut microbiota-immune-brain interactions. Clin. Nutr. 2020, 39, 676684,  DOI: 10.1016/j.clnu.2019.03.023
  161. 162
    Zhang, L.; Wang, Y.; Xiayu, X.; Shi, C.; Chen, W.; Song, N.; Fu, X.; Zhou, R.; Xu, Y. F.; Huang, L. Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2017, 60, 12411257,  DOI: 10.3233/JAD-170020
  162. 163
    Wang, L.; Christophersen, C. T.; Sorich, M. J.; Gerber, J. P.; Angley, M. T.; Conlon, M. A. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 2012, 57, 20962102,  DOI: 10.1007/s10620-012-2167-7
  163. 164
    Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 12611272,  DOI: 10.1089/jmf.2014.7000
  164. 165
    Heneka, M. T.; Carson, M. J.; El Khoury, J.; Landreth, G. E.; Brosseron, F.; Feinstein, D. L.; Jacobs, A. H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R. M. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388405,  DOI: 10.1016/S1474-4422(15)70016-5
  165. 166
    Ransohoff, R. M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777783,  DOI: 10.1126/science.aag2590
  166. 167
    Karlsson, O.; Roman, E.; Berg, A. L.; Brittebo, E. B. Early hippocampal cell death, and late learning and memory deficits in rats exposed to the environmental toxin BMAA (β-N-methylamino-L-alanine) during the neonatal period. Behav. Brain Res. 2011, 219, 310320,  DOI: 10.1016/j.bbr.2011.01.056
  167. 168
    Nunes-Costa, D.; Magalhães, J. D.; G-Fernandes, M.; Cardoso, S. M.; Empadinhas, N. Microbial BMAA and the Pathway for Parkinson’s Disease Neurodegeneration. Front. Aging Neurosci. 2020, 12, 26,  DOI: 10.3389/fnagi.2020.00026
  168. 169
    Braak, H.; Rüb, U.; Gai, W. P.; Del Tredici, K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm (Vienna) 2003, 110, 517536,  DOI: 10.1007/s00702-002-0808-2
  169. 170
    Wallen, Z. D.; Demirkan, A.; Twa, G.; Cohen, G.; Dean, M. N.; Standaert, D. G.; Sampson, T. R.; Payami, H. Metagenomics of Parkinson’s disease implicates the gut microbiome in multiple disease mechanisms. Nat. Commun. 2022, 13, 6958,  DOI: 10.1038/s41467-022-34667-x
  170. 171
    Vogt, N. M.; Kerby, R. L.; Dill-McFarland, K. A.; Harding, S. J.; Merluzzi, A. P.; Johnson, S. C.; Carlsson, C. M.; Asthana, S.; Zetterberg, H.; Blennow, K. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537,  DOI: 10.1038/s41598-017-13601-y
  171. 172
    Zhuang, Z. Q.; Shen, L. L.; Li, W. W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y. L. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2018, 63, 13371346,  DOI: 10.3233/JAD-180176
  172. 173
    Manderino, L.; Carroll, I.; Azcarate-Peril, M. A.; Rochette, A.; Heinberg, L.; Peat, C.; Steffen, K.; Mitchell, J.; Gunstad, J. Preliminary Evidence for an Association Between the Composition of the Gut Microbiome and Cognitive Function in Neurologically Healthy Older Adults. J. Int. Neuropsychol. Soc. 2017, 23, 700705,  DOI: 10.1017/S1355617717000492
  173. 174
    Barbosa, R. S. D.; Vieira-Coelho, M. A. Probiotics and prebiotics: focus on psychiatric disorders - a systematic review. Nutr. Rev. 2020, 78, 437450,  DOI: 10.1093/nutrit/nuz080
  174. 175
    Ansari, F.; Pourjafar, H.; Tabrizi, A.; Homayouni, A. The Effects of Probiotics and Prebiotics on Mental Disorders: A Review on Depression, Anxiety, Alzheimer, and Autism Spectrum Disorders. Curr. Pharm. Biotechnol. 2020, 21, 555565,  DOI: 10.2174/1389201021666200107113812
  175. 176
    Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J. F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755764,  DOI: 10.1017/S0007114510004319
  176. 177
    Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; Bahmani, F.; Kouchaki, E.; Tamtaji, O. R.; Hamidi, G. A.; Salami, M. Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2016, 8, 256,  DOI: 10.3389/fnagi.2016.00256
  177. 178
    Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, Kc01kc05,  DOI: 10.7860/JCDR/2017/26106.10428
  178. 179
    Tamtaji, O. R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 10311035,  DOI: 10.1016/j.clnu.2018.05.018
  179. 180
    Frazier, K.; Chang, E. B. Intersection of the Gut Microbiome and Circadian Rhythms in Metabolism. Trends Endocrinol Metab 2020, 31, 2536,  DOI: 10.1016/j.tem.2019.08.013
  180. 181
    Gutierrez Lopez, D. E.; Lashinger, L. M.; Weinstock, G. M.; Bray, M. S. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021, 33, 873887,  DOI: 10.1016/j.cmet.2021.03.015
  181. 182
    Leng, Y.; Musiek, E. S.; Hu, K.; Cappuccio, F. P.; Yaffe, K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol. 2019, 18, 307318,  DOI: 10.1016/S1474-4422(18)30461-7
  182. 183
    Leone, V.; Gibbons, S. M.; Martinez, K.; Hutchison, A. L.; Huang, E. Y.; Cham, C. M.; Pierre, J. F.; Heneghan, A. F.; Nadimpalli, A.; Hubert, N. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe 2015, 17, 681689,  DOI: 10.1016/j.chom.2015.03.006
  183. 184
    Nicholson, J. K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 12621267,  DOI: 10.1126/science.1223813
  184. 185
    Moran, C. P.; Shanahan, F. Gut microbiota and obesity: role in aetiology and potential therapeutic target. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 585597,  DOI: 10.1016/j.bpg.2014.07.005
  185. 186
    Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 5560,  DOI: 10.1038/nature11450
  186. 187
    Donohoe, D. R.; Wali, A.; Brylawski, B. P.; Bultman, S. J. Microbial regulation of glucose metabolism and cell-cycle progression in mammalian colonocytes. PLoS One 2012, 7, e46589,  DOI: 10.1371/journal.pone.0046589
  187. 188
    Donohoe, D. R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T. M.; Bunger, M. K.; Bultman, S. J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517526,  DOI: 10.1016/j.cmet.2011.02.018
  188. 189
    Tolhurst, G.; Heffron, H.; Lam, Y. S.; Parker, H. E.; Habib, A. M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F. M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364371,  DOI: 10.2337/db11-1019
  189. 190
    de Vos, W. M.; Tilg, H.; Van Hul, M.; Cani, P. D. Gut microbiome and health: mechanistic insights. Gut 2022, 71, 10201032,  DOI: 10.1136/gutjnl-2021-326789
  190. 191
    Yeoh, Y. K.; Zuo, T.; Lui, G. C.-Y.; Zhang, F.; Liu, Q.; Li, A. Y.; Chung, A. C.; Cheung, C. P.; Tso, E. Y.; Fung, K. S. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698706,  DOI: 10.1136/gutjnl-2020-323020
  191. 192
    Burchill, E.; Lymberopoulos, E.; Menozzi, E.; Budhdeo, S.; McIlroy, J. R.; Macnaughtan, J.; Sharma, N. The Unique Impact of COVID-19 on Human Gut Microbiome Research. Frontiers in Medicine 2021, 8, 652464,  DOI: 10.3389/fmed.2021.652464
  192. 193
    Bernard-Raichon, L.; Venzon, M.; Klein, J.; Axelrad, J. E.; Zhang, C.; Sullivan, A. P.; Hussey, G. A.; Casanovas-Massana, A.; Noval, M. G.; Valero-Jimenez, A. M. Gut microbiome dysbiosis in antibiotic-treated COVID-19 patients is associated with microbial translocation and bacteremia. Nat. Commun. 2022, 13, 5926,  DOI: 10.1038/s41467-022-33395-6
  193. 194
    Chumpitazi, B. P. The gut microbiome as a predictor of low fermentable oligosaccharides disaccharides monosaccharides and polyols diet efficacy in functional bowel disorders. Curr. Opin. Gastroenterol. 2020, 36, 147154,  DOI: 10.1097/MOG.0000000000000608
  194. 195
    Leshem, A.; Segal, E.; Elinav, E. The Gut Microbiome and Individual-Specific Responses to Diet. mSystems 2020, 5, e0066500620,  DOI: 10.1128/mSystems.00665-20
  195. 196
    Lepage, P.; Häsler, R.; Spehlmann, M. E.; Rehman, A.; Zvirbliene, A.; Begun, A.; Ott, S.; Kupcinskas, L.; Doré, J.; Raedler, A. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 2011, 141, 227236,  DOI: 10.1053/j.gastro.2011.04.011
  196. 197
    Manichanh, C.; Borruel, N.; Casellas, F.; Guarner, F. The gut microbiota in IBD. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 599608,  DOI: 10.1038/nrgastro.2012.152
  197. 198
    Tong, M.; Li, X.; Wegener Parfrey, L.; Roth, B.; Ippoliti, A.; Wei, B.; Borneman, J.; McGovern, D. P.; Frank, D. N.; Li, E. A modular organization of the human intestinal mucosal microbiota and its association with inflammatory bowel disease. PLoS One 2013, 8, e80702,  DOI: 10.1371/journal.pone.0080702
  198. 199
    Guinane, C. M.; Cotter, P. D. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap. Adv. Gastroenterol. 2013, 6, 295308,  DOI: 10.1177/1756283X13482996
  199. 200
    Kassam, Z.; Lee, C. H.; Yuan, Y.; Hunt, R. H. Fecal microbiota transplantation for Clostridium difficile infection: systematic review and meta-analysis. Am. J. Gastroenterol. 2013, 108, 500508,  DOI: 10.1038/ajg.2013.59
  200. 201
    Tang, W. H.; Kitai, T.; Hazen, S. L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 11831196,  DOI: 10.1161/CIRCRESAHA.117.309715
  201. 202
    Jiang, H. Y.; Zhang, X.; Yu, Z. H.; Zhang, Z.; Deng, M.; Zhao, J. H.; Ruan, B. Altered gut microbiota profile in patients with generalized anxiety disorder. J. Psychiatr. Res. 2018, 104, 130136,  DOI: 10.1016/j.jpsychires.2018.07.007
  202. 203
    Chen, Y. H.; Bai, J.; Wu, D.; Yu, S. F.; Qiang, X. L.; Bai, H.; Wang, H. N.; Peng, Z. W. Association between fecal microbiota and generalized anxiety disorder: Severity and early treatment response. J. Affect. Disord. 2019, 259, 5666,  DOI: 10.1016/j.jad.2019.08.014
  203. 204
    Hemmings, S. M. J.; Malan-Müller, S.; van den Heuvel, L. L.; Demmitt, B. A.; Stanislawski, M. A.; Smith, D. G.; Bohr, A. D.; Stamper, C. E.; Hyde, E. R.; Morton, J. T. The Microbiome in Posttraumatic Stress Disorder and Trauma-Exposed Controls: An Exploratory Study. Psychosom. Med. 2017, 79, 936946,  DOI: 10.1097/PSY.0000000000000512
  204. 205
    Stevens, B. R.; Goel, R.; Seungbum, K.; Richards, E. M.; Holbert, R. C.; Pepine, C. J.; Raizada, M. K. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut 2018, 67, 15551557,  DOI: 10.1136/gutjnl-2017-314759
  205. 206
    Kelly, J. R.; Borre, Y.; O'Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P. J.; Beers, S.; Scott, K.; Moloney, G. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109118,  DOI: 10.1016/j.jpsychires.2016.07.019
  206. 207
    Lin, P.; Ding, B.; Feng, C.; Yin, S.; Zhang, T.; Qi, X.; Lv, H.; Guo, X.; Dong, K.; Zhu, Y. Prevotella and Klebsiella proportions in fecal microbial communities are potential characteristic parameters for patients with major depressive disorder. J. Affect. Disord. 2017, 207, 300304,  DOI: 10.1016/j.jad.2016.09.051
  207. 208
    Gough, E.; Shaikh, H.; Manges, A. R. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin. Infect. Dis. 2011, 53, 9941002,  DOI: 10.1093/cid/cir632
  208. 209
    Craciun, C. I.; Neag, M. A.; Catinean, A.; Mitre, A. O.; Rusu, A.; Bala, C.; Roman, G.; Buzoianu, A. D.; Muntean, D. M.; Craciun, A. E. The Relationships between Gut Microbiota and Diabetes Mellitus, and Treatments for Diabetes Mellitus. Biomedicines 2022, 10, 308,  DOI: 10.3390/biomedicines10020308
  209. 210
    van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E. G.; de Vos, W. M.; Visser, C. E.; Kuijper, E. J.; Bartelsman, J. F. W. M.; Tijssen, J. G. P. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. New England Journal of Medicine 2013, 368, 407415,  DOI: 10.1056/NEJMoa1205037
  210. 211
    Hamilton, M. J.; Weingarden, A. R.; Sadowsky, M. J.; Khoruts, A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 761767,  DOI: 10.1038/ajg.2011.482
  211. 212
    Zafar, H.; Saier, M. H., Jr. Gut Bacteroides species in health and disease. Gut Microbes 2021, 13, 1848158,  DOI: 10.1080/19490976.2020.1848158
  212. 213
    Chen, Y.; Yang, Y.; Gu, J. Clinical Implications of the Associations Between Intestinal Microbiome and Colorectal Cancer Progression. Cancer Manag. Res. 2020, 12, 41174128,  DOI: 10.2147/CMAR.S240108
  213. 214
    Bullman, S.; Pedamallu, C. S.; Sicinska, E.; Clancy, T. E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 14431448,  DOI: 10.1126/science.aal5240
  214. 215
    Cammann, D.; Lu, Y.; Cummings, M. J.; Zhang, M. L.; Cue, J. M.; Do, J.; Ebersole, J.; Chen, X.; Oh, E. C.; Cummings, J. L. Genetic correlations between Alzheimer’s disease and gut microbiome genera. Sci. Rep. 2023, 13, 5258,  DOI: 10.1038/s41598-023-31730-5
  215. 216
    Silva, Y. P.; Bernardi, A.; Frozza, R. L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25,  DOI: 10.3389/fendo.2020.00025
  216. 217
    Manning, L. P.; Yao, C. K.; Biesiekierski, J. R. Therapy of IBS: Is a Low FODMAP Diet the Answer?. Front. Psychiatry 2020, 11, 865,  DOI: 10.3389/fpsyt.2020.00865
  217. 218
    Nanayakkara, W. S.; Skidmore, P. M.; O’Brien, L.; Wilkinson, T. J.; Gearry, R. B. Efficacy of the low FODMAP diet for treating irritable bowel syndrome: the evidence to date. Clin. Exp. Gastroenterol. 2016, 9, 131142,  DOI: 10.2147/CEG.S86798
  218. 219
    Tegegne, B. A.; Kebede, B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022, 8, e09725  DOI: 10.1016/j.heliyon.2022.e09725
  219. 220
    Aponte, M.; Murru, N.; Shoukat, M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front. Microbiol. 2020, 11, 562048,  DOI: 10.3389/fmicb.2020.562048
  220. 221
    Noonan, S.; Zaveri, M.; Macaninch, E.; Martyn, K. Food & mood: a review of supplementary prebiotic and probiotic interventions in the treatment of anxiety and depression in adults. BMJ. Nutrition, Prevention & Health 2020, 3, 351,  DOI: 10.1136/bmjnph-2019-000053
  221. 222
    MACFARLANE, S.; MACFARLANE, G. T.; CUMMINGS, J. H. Review article: prebiotics in the gastrointestinal tract. Aliment. Pharmacol. Ther. 2006, 24, 701714,  DOI: 10.1111/j.1365-2036.2006.03042.x
  222. 223
    Dinan, K.; Dinan, T. Antibiotics and mental health: The good, the bad and the ugly. J. Int. Med. 2022, 292, 858869,  DOI: 10.1111/joim.13543
  223. 224
    Karakan, T.; Ozkul, C.; Küpeli Akkol, E.; Bilici, S.; Sobarzo-Sánchez, E.; Capasso, R. Gut-Brain-Microbiota Axis: Antibiotics and Functional Gastrointestinal Disorders. Nutrients 2021, 13, 389,  DOI: 10.3390/nu13020389
  224. 225
    Xu, H. M.; Huang, H. L.; Zhou, Y. L.; Zhao, H. L.; Xu, J.; Shou, D. W.; Liu, Y. D.; Zhou, Y. J.; Nie, Y. Q. Fecal Microbiota Transplantation: A New Therapeutic Attempt from the Gut to the Brain. Gastroenterol. Res. Pract. 2021, 2021, 6699268,  DOI: 10.1155/2021/6699268
  225. 226
    Doll, J. P. K.; Vázquez-Castellanos, J. F.; Schaub, A.-C.; Schweinfurth, N.; Kettelhack, C.; Schneider, E.; Yamanbaeva, G.; Mählmann, L.; Brand, S.; Beglinger, C. Fecal Microbiota Transplantation (FMT) as an Adjunctive Therapy for Depression─Case Report. Front. Psychiatry 2022, 13, 815422,  DOI: 10.3389/fpsyt.2022.815422
  226. 227
    Waller, K. M. J.; Leong, R. W.; Paramsothy, S. An update on fecal microbiota transplantation for the treatment of gastrointestinal diseases. J. Gastroenterol. Hepatol. 2022, 37, 246255,  DOI: 10.1111/jgh.15731
  227. 228
    Hetterich, L.; Stengel, A. Psychotherapeutic Interventions in Irritable Bowel Syndrome. Front. Psychiatry 2020, 11, 286,  DOI: 10.3389/fpsyt.2020.00286
  228. 229
    Wildes, J. E.; Bedell, A.; Graham, A. K.; Kells, M. Brain-gut psychotherapies: Promising tools to address gastrointestinal problems in patients with eating disorders. Int. J. Eat. Disord. 2021, 54, 10631067,  DOI: 10.1002/eat.23555
  229. 230
    Laird, K. T.; Tanner-Smith, E. E.; Russell, A. C.; Hollon, S. D.; Walker, L. S. Comparative efficacy of psychological therapies for improving mental health and daily functioning in irritable bowel syndrome: A systematic review and meta-analysis. Clin. Psychol. Rev. 2017, 51, 142152,  DOI: 10.1016/j.cpr.2016.11.001
  230. 231
    Colomier, E.; Algera, J.; Melchior, C. Pharmacological Therapies and Their Clinical Targets in Irritable Bowel Syndrome With Diarrhea. Front. Pharmacol. 2021, 11, 629026,  DOI: 10.3389/fphar.2020.629026
  231. 232
    World Health Organization. Pharmacological treatment of mental disorders in primary health care. WHO Press: Geneva, Switzerland, 2009.
  232. 233
    Fikree, A.; Byrne, P. Management of functional gastrointestinal disorders. Clin Med. (Lond) 2021, 21, 4452,  DOI: 10.7861/clinmed.2020-0980
  233. 234
    Tierney, B. T.; Yang, Z.; Luber, J. M.; Beaudin, M.; Wibowo, M. C.; Baek, C.; Mehlenbacher, E.; Patel, C. J.; Kostic, A. D. The Landscape of Genetic Content in the Gut and Oral Human Microbiome. Cell Host Microbe 2019, 26, 283295.e288,  DOI: 10.1016/j.chom.2019.07.008
  234. 235
    International Human Genome Sequencing Consortium Finishing the euchromatic sequence of the human genome. Nature 2004, 431, 931945,  DOI: 10.1038/nature03001
  235. 236
    Liu, J.; Tan, Y.; Cheng, H.; Zhang, D.; Feng, W.; Peng, C. Functions of Gut Microbiota Metabolites, Current Status and Future Perspectives. Aging Dis. 2022, 13, 11061126,  DOI: 10.14336/AD.2022.0104
  236. 237
    Heijtz, R. D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M. L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 30473052,  DOI: 10.1073/pnas.1010529108
  237. 238
    Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.-N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. Journal of Physiology 2004, 558, 263275,  DOI: 10.1113/jphysiol.2004.063388
  238. 239
    Needham, B. D.; Funabashi, M.; Adame, M. D.; Wang, Z.; Boktor, J. C.; Haney, J.; Wu, W.-L.; Rabut, C.; Ladinsky, M. S.; Hwang, S.-J. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647653,  DOI: 10.1038/s41586-022-04396-8
  239. 240
    Gacias, M.; Gaspari, S.; Santos, P.-M. G.; Tamburini, S.; Andrade, M.; Zhang, F.; Shen, N.; Tolstikov, V.; Kiebish, M. A.; Dupree, J. L. Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior. eLife 2016, 5, e13442,  DOI: 10.7554/eLife.13442
  240. 241
    Vuong, H. E.; Pronovost, G. N.; Williams, D. W.; Coley, E. J. L.; Siegler, E. L.; Qiu, A.; Kazantsev, M.; Wilson, C. J.; Rendon, T.; Hsiao, E. Y. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020, 586, 281286,  DOI: 10.1038/s41586-020-2745-3
  241. 242
    Wei, G. Z.; Martin, K. A.; Xing, P. Y.; Agrawal, R.; Whiley, L.; Wood, T. K.; Hejndorf, S.; Ng, Y. Z.; Low, J. Z. Y.; Rossant, J. Tryptophan-metabolizing gut microbes regulate adult neurogenesis via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. U. S. A. 2021, 118, e2021091118,  DOI: 10.1073/pnas.2021091118
  242. 243
    Kang, D.-W.; Adams, J. B.; Gregory, A. C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 2017, 5, 10,  DOI: 10.1186/s40168-016-0225-7
  243. 244
    Korpela, K.; Helve, O.; Kolho, K. L.; Saisto, T.; Skogberg, K.; Dikareva, E.; Stefanovic, V.; Salonen, A.; Andersson, S.; de Vos, W. M. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell 2020, 183, 324334.e325,  DOI: 10.1016/j.cell.2020.08.047
  244. 245
    Wikoff, W. R.; Anfora, A. T.; Liu, J.; Schultz, P. G.; Lesley, S. A.; Peters, E. C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 36983703,  DOI: 10.1073/pnas.0812874106
  245. 246
    Dodd, D.; Spitzer, M. H.; Van Treuren, W.; Merrill, B. D.; Hryckowian, A. J.; Higginbottom, S. K.; Le, A.; Cowan, T. M.; Nolan, G. P.; Fischbach, M. A. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648652,  DOI: 10.1038/nature24661
  246. 247
    Barrett, E.; Ross, R. P.; O’Toole, P. W.; Fitzgerald, G. F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411417,  DOI: 10.1111/j.1365-2672.2012.05344.x
  247. 248
    Asano, Y.; Hiramoto, T.; Nishino, R.; Aiba, Y.; Kimura, T.; Yoshihara, K.; Koga, Y.; Sudo, N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G12881295,  DOI: 10.1152/ajpgi.00341.2012
  248. 249
    Pocivavsek, A.; Wu, H.-Q.; Potter, M. C.; Elmer, G. I.; Pellicciari, R.; Schwarcz, R. Fluctuations in Endogenous Kynurenic Acid Control Hippocampal Glutamate and Memory. Neuropsychopharmacology 2011, 36, 23572367,  DOI: 10.1038/npp.2011.127
  249. 250
    Potter, M. C.; Elmer, G. I.; Bergeron, R.; Albuquerque, E. X.; Guidetti, P.; Wu, H.-Q.; Schwarcz, R. Reduction of Endogenous Kynurenic Acid Formation Enhances Extracellular Glutamate, Hippocampal Plasticity, and Cognitive Behavior. Neuropsychopharmacology 2010, 35, 17341742,  DOI: 10.1038/npp.2010.39
  250. 251
    Reigstad, C. S.; Salmonson, C. E.; Rainey, J. F., III; Szurszewski, J. H.; Linden, D. R.; Sonnenburg, J. L.; Farrugia, G.; Kashyap, P. C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 13951403,  DOI: 10.1096/fj.14-259598
  251. 252
    Yano, J. M.; Yu, K.; Donaldson, G. P.; Shastri, G. G.; Ann, P.; Ma, L.; Nagler, C. R.; Ismagilov, R. F.; Mazmanian, S. K.; Hsiao, E. Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264276,  DOI: 10.1016/j.cell.2015.02.047
  252. 253
    Bellono, N. W.; Bayrer, J. R.; Leitch, D. B.; Castro, J.; Zhang, C.; O’Donnell, T. A.; Brierley, S. M.; Ingraham, H. A.; Julius, D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017, 170, 185198.e16,  DOI: 10.1016/j.cell.2017.05.034
  253. 254
    Ye, L.; Bae, M.; Cassilly, C. D.; Jabba, S. V.; Thorpe, D. W.; Martin, A. M.; Lu, H. Y.; Wang, J.; Thompson, J. D.; Lickwar, C. R. Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways. Cell Host Microbe 2021, 29, 179196.e79,  DOI: 10.1016/j.chom.2020.11.011
  254. 255
    O’Hagan, C.; Li, J. V.; Marchesi, J. R.; Plummer, S.; Garaiova, I.; Good, M. A. Long-term multi-species Lactobacillus and Bifidobacterium dietary supplement enhances memory and changes regional brain metabolites in middle-aged rats. Neurobiol Learn Mem 2017, 144, 3647,  DOI: 10.1016/j.nlm.2017.05.015
  255. 256
    Mao, J.-H.; Kim, Y.-M.; Zhou, Y.-X.; Hu, D.; Zhong, C.; Chang, H.; Brislawn, C. J.; Fansler, S.; Langley, S.; Wang, Y. Genetic and metabolic links between the murine microbiome and memory. Microbiome 2020, 8, 53,  DOI: 10.1186/s40168-020-00817-w
  256. 257
    El Hayek, L.; Khalifeh, M.; Zibara, V.; Abi Assaad, R.; Emmanuel, N.; Karnib, N.; El-Ghandour, R.; Nasrallah, P.; Bilen, M.; Ibrahim, P. Lactate Mediates the Effects of Exercise on Learning and Memory through SIRT1-Dependent Activation of Hippocampal Brain-Derived Neurotrophic Factor (BDNF). J. Neurosci. 2019, 39, 23692382,  DOI: 10.1523/JNEUROSCI.1661-18.2019
  257. 258
    Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nature Reviews Gastroenterology & Hepatology 2019, 16, 461478,  DOI: 10.1038/s41575-019-0157-3
  258. 259
    van de Wouw, M.; Boehme, M.; Lyte, J. M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain–gut axis alterations. Journal of Physiology 2018, 596, 49234944,  DOI: 10.1113/JP276431
  259. 260
    Bernasconi, R.; Jones, R. S.; Bittiger, H.; Olpe, H. R.; Heid, J.; Martin, P.; Klein, M.; Loo, P.; Braunwalder, A.; Schmutz, M. Dose pipecolic acid interact with the central GABA-ergic system?. J. Neural Transm. 1986, 67, 175189,  DOI: 10.1007/BF01243346
  260. 261
    Takahama, K.; Hashimoto, T.; Wang, M. W.; Akaike, N.; Hitoshi, T.; Okano, Y.; Kasé, Y.; Miyata, T. Pipecolic acid enhancement of GABA response in single neurons of rat brain. Neuropharmacology 1986, 25, 339342,  DOI: 10.1016/0028-3908(86)90263-7
  261. 262
    Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L. G.; Kundu, P. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158263ra158,  DOI: 10.1126/scitranslmed.3009759
  262. 263
    Li, H.; Sun, J.; Wang, F.; Ding, G.; Chen, W.; Fang, R.; Yao, Y.; Pang, M.; Lu, Z. Q.; Liu, J. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016, 1642, 7078,  DOI: 10.1016/j.brainres.2016.03.031
  263. 264
    Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J. K.; Carding, S. R.; Glen, R. C.; McArthur, S. Microbiome–host systems interactions: protective effects of propionate upon the blood–brain barrier. Microbiome 2018, 6, 55,  DOI: 10.1186/s40168-018-0439-y
  264. 265
    Quinn, M.; McMillin, M.; Galindo, C.; Frampton, G.; Pae, H. Y.; DeMorrow, S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig. Liver Dis. 2014, 46, 527534,  DOI: 10.1016/j.dld.2014.01.159
  265. 266
    Palmela, I.; Correia, L.; Silva, R. F. M.; Sasaki, H.; Kim, K. S.; Brites, D.; Brito, M. A. Hydrophilic bile acids protect human blood-brain barrier endothelial cells from disruption by unconjugated bilirubin: an in vitro study. Front. Neurosci. 2015, 9, 80,  DOI: 10.3389/fnins.2015.00080
  266. 267
    Hoyles, L.; Pontifex, M. G.; Rodriguez-Ramiro, I.; Anis-Alavi, M. A.; Jelane, K. S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A. L.; Carding, S. R. Regulation of blood–brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235,  DOI: 10.1186/s40168-021-01181-z
  267. 268
    Magistretti, P. J.; Allaman, I. A cellular perspective on brain energy metabolism and functional imaging. Neuron 2015, 86, 883901,  DOI: 10.1016/j.neuron.2015.03.035
  268. 269
    Wyss, M. T.; Jolivet, R.; Buck, A.; Magistretti, P. J.; Weber, B. In Vivo Evidence for Lactate as a Neuronal Energy Source. J. Neurosci. 2011, 31, 74777485,  DOI: 10.1523/JNEUROSCI.0415-11.2011
  269. 270
    Liu, Z.; Dai, X.; Zhang, H.; Shi, R.; Hui, Y.; Jin, X.; Zhang, W.; Wang, L.; Wang, Q.; Wang, D. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat. Commun. 2020, 11, 855,  DOI: 10.1038/s41467-020-14676-4
  270. 271
    Russell, W. R.; Hoyles, L.; Flint, H. J.; Dumas, M. E. Colonic bacterial metabolites and human health. Curr. Opin. Microbiol. 2013, 16, 246254,  DOI: 10.1016/j.mib.2013.07.002
  271. 272
    Ren, Z.; Zhang, R.; Li, Y.; Li, Y.; Yang, Z.; Yang, H. Ferulic acid exerts neuroprotective effects against cerebral ischemia/reperfusion-induced injury via antioxidant and anti-apoptotic mechanisms in vitro and in vivo. Int. J. Mol. Med. 2017, 40, 14441456,  DOI: 10.3892/ijmm.2017.3127
  272. 273
    Zeni, A. L. B.; Camargo, A.; Dalmagro, A. P. Ferulic acid reverses depression-like behavior and oxidative stress induced by chronic corticosterone treatment in mice. Steroids 2017, 125, 131136,  DOI: 10.1016/j.steroids.2017.07.006
  273. 274
    Verzelloni, E.; Pellacani, C.; Tagliazucchi, D.; Tagliaferri, S.; Calani, L.; Costa, L. G.; Brighenti, F.; Borges, G.; Crozier, A.; Conte, A. Antiglycative and neuroprotective activity of colon-derived polyphenol catabolites. Mol. Nutr. Food Res. 2011, 55, S35S43,  DOI: 10.1002/mnfr.201000525
  274. 275
    Macfarlane, S.; Macfarlane, G. T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 6772,  DOI: 10.1079/PNS2002207
  275. 276
    Thomas, C. M.; Hong, T.; van Pijkeren, J. P.; Hemarajata, P.; Trinh, D. V.; Hu, W.; Britton, R. A.; Kalkum, M.; Versalovic, J. Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PLoS One 2012, 7, e31951,  DOI: 10.1371/journal.pone.0031951
  276. 277
    De Biase, D.; Pennacchietti, E. Glutamate decarboxylase-dependent acid resistance in orally acquired bacteria: function, distribution and biomedical implications of the gadBC operon. Mol. Microbiol. 2012, 86, 770786,  DOI: 10.1111/mmi.12020
  277. 278
    Zhou, L.; Zeng, Y.; Zhang, H.; Ma, Y. The Role of Gastrointestinal Microbiota in Functional Dyspepsia: A Review. Front. Physiol. 2022, 13, 910568,  DOI: 10.3389/fphys.2022.910568
  278. 279
    Alexandrov, P. N.; Hill, J. M.; Zhao, Y.; Bond, T.; Taylor, C. M.; Percy, M. E.; Li, W.; Lukiw, W. J. Aluminum-induced generation of lipopolysaccharide (LPS) from the human gastrointestinal (GI)-tract microbiome-resident Bacteroides fragilis. J. Inorg. Biochem. 2020, 203, 110886,  DOI: 10.1016/j.jinorgbio.2019.110886
  279. 280
    Brown, E. M.; Ke, X.; Hitchcock, D.; Jeanfavre, S.; Avila-Pacheco, J.; Nakata, T.; Arthur, T. D.; Fornelos, N.; Heim, C.; Franzosa, E. A. Bacteroides-Derived Sphingolipids Are Critical for Maintaining Intestinal Homeostasis and Symbiosis. Cell Host Microbe 2019, 25, 668680.e67,  DOI: 10.1016/j.chom.2019.04.002
  280. 281
    Gao, K.; Pi, Y.; Mu, C. L.; Farzi, A.; Liu, Z.; Zhu, W. Y. Increasing carbohydrate availability in the hindgut promotes hypothalamic neurotransmitter synthesis: aromatic amino acids linking the microbiota-brain axis. J. Neurochem. 2019, 149, 641659,  DOI: 10.1111/jnc.14709
  281. 282
    Gao, K.; Mu, C.-l.; Farzi, A.; Zhu, W.-y. Tryptophan Metabolism: A Link Between the Gut Microbiota and Brain. Adv. Nutr. 2020, 11, 709723,  DOI: 10.1093/advances/nmz127
  282. 283
    Xiao, L.; Liu, Q.; Luo, M.; Xiong, L. Gut Microbiota-Derived Metabolites in Irritable Bowel Syndrome. Frontiers in Cellular and Infection Microbiology 2021, 11, 729346,  DOI: 10.3389/fcimb.2021.729346
  283. 284
    Mawe, G. M.; Hoffman, J. M. Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473486,  DOI: 10.1038/nrgastro.2013.105
  284. 285
    Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A. N.; Mackay, C. R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91119,  DOI: 10.1016/B978-0-12-800100-4.00003-9
  285. 286
    Felizardo, R. J. F.; Watanabe, I. K. M.; Dardi, P.; Rossoni, L. V.; Câmara, N. O. S. The interplay among gut microbiota, hypertension and kidney diseases: The role of short-chain fatty acids. Pharmacol. Res. 2019, 141, 366377,  DOI: 10.1016/j.phrs.2019.01.019
  286. 287
    Canfora, E. E.; Meex, R. C. R.; Venema, K.; Blaak, E. E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 2019, 15, 261273,  DOI: 10.1038/s41574-019-0156-z
  287. 288
    Tahara, Y.; Yamazaki, M.; Sukigara, H.; Motohashi, H.; Sasaki, H.; Miyakawa, H.; Haraguchi, A.; Ikeda, Y.; Fukuda, S.; Shibata, S. Gut Microbiota-Derived Short Chain Fatty Acids Induce Circadian Clock Entrainment in Mouse Peripheral Tissue. Sci. Rep. 2018, 8, 1395,  DOI: 10.1038/s41598-018-19836-7
  288. 289
    Feng, W.; Ao, H.; Peng, C. Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Front. Pharmacol. 2018, 9, 1354,  DOI: 10.3389/fphar.2018.01354
  289. 290
    Pan, L. L.; Li, B. B.; Pan, X. H.; Sun, J. Gut microbiota in pancreatic diseases: possible new therapeutic strategies. Acta Pharmacol. Sin. 2021, 42, 10271039,  DOI: 10.1038/s41401-020-00532-0
  290. 291
    Perino, A.; Demagny, H.; Velazquez-Villegas, L.; Schoonjans, K. Molecular Physiology of Bile Acid Signaling in Health, Disease, and Aging. Physiol. Rev. 2021, 101, 683731,  DOI: 10.1152/physrev.00049.2019
  291. 292
    Wahlström, A.; Sayin, S. I.; Marschall, H. U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 4150,  DOI: 10.1016/j.cmet.2016.05.005
  292. 293
    Poland, J. C.; Flynn, C. R. Bile Acids, Their Receptors, and the Gut Microbiota. Physiology (Bethesda) 2021, 36, 235245,  DOI: 10.1152/physiol.00028.2020
  293. 294
    Govindarajan, K.; MacSharry, J.; Casey, P. G.; Shanahan, F.; Joyce, S. A.; Gahan, C. G. M. Unconjugated Bile Acids Influence Expression of Circadian Genes: A Potential Mechanism for Microbe-Host Crosstalk. PLoS One 2016, 11, e0167319  DOI: 10.1371/journal.pone.0167319
  294. 295
    McMillin, M.; DeMorrow, S. Effects of bile acids on neurological function and disease. FASEB J. 2016, 30, 36583668,  DOI: 10.1096/fj.201600275R
  295. 296
    Roth, W.; Zadeh, K.; Vekariya, R.; Ge, Y.; Mohamadzadeh, M. Tryptophan Metabolism and Gut-Brain Homeostasis. Int. J. Mol. Sci. 2021, 22, 2973,  DOI: 10.3390/ijms22062973
  296. 297
    Zhang, J.; Zhu, S.; Ma, N.; Johnston, L. J.; Wu, C.; Ma, X. Metabolites of microbiota response to tryptophan and intestinal mucosal immunity: A therapeutic target to control intestinal inflammation. Med. Res. Rev. 2021, 41, 10611088,  DOI: 10.1002/med.21752
  297. 298
    Modoux, M.; Rolhion, N.; Mani, S.; Sokol, H. Tryptophan Metabolism as a Pharmacological Target. Trends Pharmacol. Sci. 2021, 42, 6073,  DOI: 10.1016/j.tips.2020.11.006
  298. 299
    Roager, H. M.; Licht, T. R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294,  DOI: 10.1038/s41467-018-05470-4
  299. 300
    Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716724,  DOI: 10.1016/j.chom.2018.05.003
  300. 301
    Wang, Z.; Bergeron, N.; Levison, B. S.; Li, X. S.; Chiu, S.; Jia, X.; Koeth, R. A.; Li, L.; Wu, Y.; Tang, W. H. W. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583594,  DOI: 10.1093/eurheartj/ehy799
  301. 302
    Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: how gut microbiota contributes to heart failure. Transl. Res. 2021, 228, 109125,  DOI: 10.1016/j.trsl.2020.08.007
  302. 303
    Yang, S.; Li, X.; Yang, F.; Zhao, R.; Pan, X.; Liang, J.; Tian, L.; Li, X.; Liu, L.; Xing, Y. Gut Microbiota-Dependent Marker TMAO in Promoting Cardiovascular Disease: Inflammation Mechanism, Clinical Prognostic, and Potential as a Therapeutic Target. Front. Pharmacol. 2019, 10, 1360,  DOI: 10.3389/fphar.2019.01360
  303. 304
    Rudzki, L.; Stone, T. W.; Maes, M.; Misiak, B.; Samochowiec, J.; Szulc, A. Gut microbiota-derived vitamins - underrated powers of a multipotent ally in psychiatric health and disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 107, 110240,  DOI: 10.1016/j.pnpbp.2020.110240
  304. 305
    Stacchiotti, V.; Rezzi, S.; Eggersdorfer, M.; Galli, F. Metabolic and functional interplay between gut microbiota and fat-soluble vitamins. Crit Rev. Food Sci. Nutr 2021, 61, 32113232,  DOI: 10.1080/10408398.2020.1793728
  305. 306
    Masri, O. A.; Chalhoub, J. M.; Sharara, A. I. Role of vitamins in gastrointestinal diseases. World J. Gastroenterol. 2015, 21, 51915209,  DOI: 10.3748/wjg.v21.i17.5191
  306. 307
    McCarville, J. L.; Chen, G. Y.; Cuevas, V. D.; Troha, K.; Ayres, J. S. Microbiota Metabolites in Health and Disease. Annu. Rev. Immunol. 2020, 38, 147170,  DOI: 10.1146/annurev-immunol-071219-125715
  307. 308
    Wang, Y.; Li, N.; Yang, J. J.; Zhao, D. M.; Chen, B.; Zhang, G. Q.; Chen, S.; Cao, R. F.; Yu, H.; Zhao, C. Y. Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacol. Res. 2020, 157, 104784,  DOI: 10.1016/j.phrs.2020.104784
  308. 309
    Morais, L. H.; Schreiber, H. L. t.; Mazmanian, S. K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241255,  DOI: 10.1038/s41579-020-00460-0
  309. 310
    Cani, P. D.; Amar, J.; Iglesias, M. A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A. M.; Fava, F.; Tuohy, K. M.; Chabo, C. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 17611772,  DOI: 10.2337/db06-1491
  310. 311
    Serino, M.; Luche, E.; Gres, S.; Baylac, A.; Bergé, M.; Cenac, C.; Waget, A.; Klopp, P.; Iacovoni, J.; Klopp, C. Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 2012, 61, 543553,  DOI: 10.1136/gutjnl-2011-301012
  311. 312
    Ostojic, S. M. Inadequate Production of H(2) by Gut Microbiota and Parkinson Disease. Trends Endocrinol Metab 2018, 29, 286288,  DOI: 10.1016/j.tem.2018.02.006
  312. 313
    Kalantar-Zadeh, K.; Berean, K. J.; Burgell, R. E.; Muir, J. G.; Gibson, P. R. Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 733747,  DOI: 10.1038/s41575-019-0193-z
  313. 314
    Pacher, P.; Beckman, J. S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315424,  DOI: 10.1152/physrev.00029.2006
  314. 315
    Singh, S. B.; Lin, H. C. Hydrogen Sulfide in Physiology and Diseases of the Digestive Tract. Microorganisms 2015, 3, 866889,  DOI: 10.3390/microorganisms3040866
  315. 316
    Sen, N. Functional and Molecular Insights of Hydrogen Sulfide Signaling and Protein Sulfhydration. J. Mol. Biol. 2017, 429, 543561,  DOI: 10.1016/j.jmb.2016.12.015
  316. 317
    Skonieczna-Żydecka, K.; Marlicz, W.; Misera, A.; Koulaouzidis, A.; Łoniewski, I. Microbiome─The Missing Link in the Gut-Brain Axis: Focus on Its Role in Gastrointestinal and Mental Health. Journal of Clinical Medicine 2018, 7, 521,  DOI: 10.3390/jcm7120521
  317. 318
    Schmulson, M. J.; Drossman, D. A. What Is New in Rome IV. J. Neurogastroenterol. Motil. 2017, 23, 151163,  DOI: 10.5056/jnm16214
  318. 319
    Stasi, C.; Nisita, C.; Cortopassi, S.; Corretti, G.; Gambaccini, D.; De Bortoli, N.; Fani, B.; Simonetti, N.; Ricchiuti, A.; Dell’Osso, L. Subthreshold Psychiatric Psychopathology in Functional Gastrointestinal Disorders: Can It Be the Bridge between Gastroenterology and Psychiatry?. Gastroenterol. Res. Pract. 2017, 2017, 1953435,  DOI: 10.1155/2017/1953435
  319. 320
    Cryan, J. F.; O’Riordan, K. J.; Cowan, C. S. M.; Sandhu, K. V.; Bastiaanssen, T. F. S.; Boehme, M.; Codagnone, M. G.; Cussotto, S.; Fulling, C.; Golubeva, A. V. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 18772013,  DOI: 10.1152/physrev.00018.2018
  320. 321
    Gralnek, I. M.; Hays, R. D.; Kilbourne, A.; Naliboff, B.; Mayer, E. A. The impact of irritable bowel syndrome on health-related quality of life. Gastroenterology 2000, 119, 654660,  DOI: 10.1053/gast.2000.16484
  321. 322
    O’Keeffe, M.; Lomer, M. C. Who should deliver the low FODMAP diet and what educational methods are optimal: a review. J. Gastroenterol. Hepatol. 2017, 32, 2326,  DOI: 10.1111/jgh.13690
  322. 323
    Koloski, N. A.; Jones, M.; Talley, N. J. Evidence that independent gut-to-brain and brain-to-gut pathways operate in the irritable bowel syndrome and functional dyspepsia: a 1-year population-based prospective study. Alimentary pharmacology & therapeutics 2016, 44, 592600,  DOI: 10.1111/apt.13738
  323. 324
    Eijsbouts, C.; Zheng, T.; Kennedy, N. A.; Bonfiglio, F.; Anderson, C. A.; Moutsianas, L.; Holliday, J.; Shi, J.; Shringarpure, S.; Agee, M. Genome-wide analysis of 53,400 people with irritable bowel syndrome highlights shared genetic pathways with mood and anxiety disorders. Nat. Genet. 2021, 53, 15431552,  DOI: 10.1038/s41588-021-00950-8
  324. 325
    Schwarz, E.; Maukonen, J.; Hyytiäinen, T.; Kieseppä, T.; Orešič, M.; Sabunciyan, S.; Mantere, O.; Saarela, M.; Yolken, R.; Suvisaari, J. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr. Res. 2018, 192, 398403,  DOI: 10.1016/j.schres.2017.04.017
  325. 326
    Castro-Nallar, E.; Bendall, M. L.; Pérez-Losada, M.; Sabuncyan, S.; Severance, E. G.; Dickerson, F. B.; Schroeder, J. R.; Yolken, R. H.; Crandall, K. A. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ. 2015, 3, e1140,  DOI: 10.7717/peerj.1140
  326. 327
    Belkaid, Y.; Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121141,  DOI: 10.1016/j.cell.2014.03.011
  327. 328
    Anderson, G.; Maes, M. Bipolar disorder: role of immune-inflammatory cytokines, oxidative and nitrosative stress and tryptophan catabolites. Curr. Psychiatry Rep 2015, 17, 8,  DOI: 10.1007/s11920-014-0541-1
  328. 329
    Clarke, G.; Fitzgerald, P.; Cryan, J. F.; Cassidy, E. M.; Quigley, E. M.; Dinan, T. G. Tryptophan degradation in irritable bowel syndrome: evidence of indoleamine 2,3-dioxygenase activation in a male cohort. BMC Gastroenterol. 2009, 9, 6,  DOI: 10.1186/1471-230X-9-6
  329. 330
    Clarke, G.; McKernan, D.; Gaszner, G.; Quigley, E.; Cryan, J.; Dinan, T. A Distinct Profile of Tryptophan Metabolism along the Kynurenine Pathway Downstream of Toll-Like Receptor Activation in Irritable Bowel Syndrome. Front. Pharmacol. 2012, 3, 90,  DOI: 10.3389/fphar.2012.00090
  330. 331
    Liang, X.; FitzGerald, G. A. Timing the Microbes: The Circadian Rhythm of the Gut Microbiome. J. Biol. Rhythms 2017, 32, 505515,  DOI: 10.1177/0748730417729066
  331. 332
    Enck, P.; Aziz, Q.; Barbara, G.; Farmer, A. D.; Fukudo, S.; Mayer, E. A.; Niesler, B.; Quigley, E. M.; Rajilić-Stojanović, M.; Schemann, M. Irritable bowel syndrome. Nat. Rev. Dis Primers 2016, 2, 16014,  DOI: 10.1038/nrdp.2016.14
  332. 333
    Dalile, B.; Vervliet, B.; Bergonzelli, G.; Verbeke, K.; Van Oudenhove, L. Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: a randomized, placebo-controlled trial. Neuropsychopharmacology 2020, 45, 22572266,  DOI: 10.1038/s41386-020-0732-x
  333. 334
    Bercik, P.; Park, A. J.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; Deng, Y.; Blennerhassett, P. A.; Fahnestock, M.; Moine, D. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol. Motil. 2011, 23, 11321139,  DOI: 10.1111/j.1365-2982.2011.01796.x
  334. 335
    Mishima, Y.; Ishihara, S. Molecular Mechanisms of Microbiota-Mediated Pathology in Irritable Bowel Syndrome. Int. J. Mol. Sci. 2020, 21, 8664,  DOI: 10.3390/ijms21228664
  335. 336
    Leeming, E. R.; Johnson, A. J.; Spector, T. D.; Le Roy, C. I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862,  DOI: 10.3390/nu11122862
  336. 337
    Hillestad, E. M. R.; van der Meeren, A.; Nagaraja, B. H.; Bjo̷rsvik, B. R.; Haleem, N.; Benitez-Paez, A.; Sanz, Y.; Hausken, T.; Lied, G. A.; Lundervold, A. Gut bless you: The microbiota-gut-brain axis in irritable bowel syndrome. World J. Gastroenterol. 2022, 28, 412431,  DOI: 10.3748/wjg.v28.i4.412
  337. 338
    Hugerth, L. W.; Andreasson, A.; Talley, N. J.; Forsberg, A. M.; Kjellström, L.; Schmidt, P. T.; Agreus, L.; Engstrand, L. No distinct microbiome signature of irritable bowel syndrome found in a Swedish random population. Gut 2020, 69, 10761084,  DOI: 10.1136/gutjnl-2019-318717
  338. 339
    Sundin, J.; Aziz, I.; Nordlander, S.; Polster, A.; Hu, Y. O. O.; Hugerth, L. W.; Pennhag, A. A. L.; Engstrand, L.; Törnblom, H.; Simrén, M. Evidence of altered mucosa-associated and fecal microbiota composition in patients with Irritable Bowel Syndrome. Sci. Rep. 2020, 10, 593,  DOI: 10.1038/s41598-020-57468-y
  339. 340
    Mayer, E. A.; Labus, J.; Aziz, Q.; Tracey, I.; Kilpatrick, L.; Elsenbruch, S.; Schweinhardt, P.; Van Oudenhove, L.; Borsook, D. Role of brain imaging in disorders of brain-gut interaction: a Rome Working Team Report. Gut 2019, 68, 17011715,  DOI: 10.1136/gutjnl-2019-318308
  340. 341
    Seminowicz, D. A.; Labus, J. S.; Bueller, J. A.; Tillisch, K.; Naliboff, B. D.; Bushnell, M. C.; Mayer, E. A. Regional gray matter density changes in brains of patients with irritable bowel syndrome. Gastroenterology 2010, 139, 4857.e42,  DOI: 10.1053/j.gastro.2010.03.049
  341. 342
    Kano, M.; Grinsvall, C.; Ran, Q.; Dupont, P.; Morishita, J.; Muratsubaki, T.; Mugikura, S.; Ly, H. G.; Törnblom, H.; Ljungberg, M. Resting state functional connectivity of the pain matrix and default mode network in irritable bowel syndrome: a graph theoretical analysis. Sci. Rep. 2020, 10, 11015,  DOI: 10.1038/s41598-020-67048-9
  342. 343
    Wilder-Smith, C. H.; Schindler, D.; Lovblad, K.; Redmond, S. M.; Nirkko, A. Brain functional magnetic resonance imaging of rectal pain and activation of endogenous inhibitory mechanisms in irritable bowel syndrome patient subgroups and healthy controls. Gut 2004, 53, 15951601,  DOI: 10.1136/gut.2003.028514
  343. 344
    Tillisch, K.; Mayer, E. A.; Labus, J. S. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology 2011, 140, 91100,  DOI: 10.1053/j.gastro.2010.07.053
  344. 345
    Ringel, Y.; Drossman, D. A.; Leserman, J. L.; Suyenobu, B. Y.; Wilber, K.; Lin, W.; Whitehead, W. E.; Naliboff, B. D.; Berman, S.; Mayer, E. A. Effect of abuse history on pain reports and brain responses to aversive visceral stimulation: an FMRI study. Gastroenterology 2008, 134, 396404,  DOI: 10.1053/j.gastro.2007.11.011
  345. 346
    Mayer, E. A.; Bradesi, S.; Chang, L.; Spiegel, B. M.; Bueller, J. A.; Naliboff, B. D. Functional GI disorders: from animal models to drug development. Gut 2008, 57, 384404,  DOI: 10.1136/gut.2006.101675
  346. 347
    Labus, J. S.; Osadchiy, V.; Hsiao, E. Y.; Tap, J.; Derrien, M.; Gupta, A.; Tillisch, K.; Le Nevé, B.; Grinsvall, C.; Ljungberg, M. Evidence for an association of gut microbial Clostridia with brain functional connectivity and gastrointestinal sensorimotor function in patients with irritable bowel syndrome, based on tripartite network analysis. Microbiome 2019, 7, 45,  DOI: 10.1186/s40168-019-0656-z
  347. 348
    Osadchiy, V.; Mayer, E. A.; Gao, K.; Labus, J. S.; Naliboff, B.; Tillisch, K.; Chang, L.; Jacobs, J. P.; Hsiao, E. Y.; Gupta, A. Analysis of brain networks and fecal metabolites reveals brain-gut alterations in premenopausal females with irritable bowel syndrome. Transl Psychiatry 2020, 10, 367,  DOI: 10.1038/s41398-020-01071-2
  348. 349
    Sarnoff, R. P.; Bhatt, R. R.; Osadchiy, V.; Dong, T.; Labus, J. S.; Kilpatrick, L. A.; Chen, Z.; Subramanyam, V.; Zhang, Y.; Ellingson, B. M. A multi-omic brain gut microbiome signature differs between IBS subjects with different bowel habits. Neuropharmacology 2023, 225, 109381,  DOI: 10.1016/j.neuropharm.2022.109381
  349. 350
    Berman, S. M.; Naliboff, B. D.; Suyenobu, B.; Labus, J. S.; Stains, J.; Ohning, G.; Kilpatrick, L.; Bueller, J. A.; Ruby, K.; Jarcho, J. Reduced brainstem inhibition during anticipated pelvic visceral pain correlates with enhanced brain response to the visceral stimulus in women with irritable bowel syndrome. J. Neurosci. 2008, 28, 349359,  DOI: 10.1523/JNEUROSCI.2500-07.2008
  350. 351
    Guleria, A.; Karyampudi, A.; Singh, R.; Khetrapal, C. L.; Verma, A.; Ghoshal, U. C.; Kumar, D. Mapping of Brain Activations to Rectal Balloon Distension Stimuli in Male Patients with Irritable Bowel Syndrome Using Functional Magnetic Resonance Imaging. J. Neurogastroenterol. Motil. 2017, 23, 415427,  DOI: 10.5056/jnm16148
  351. 352
    Ticinesi, A.; Milani, C.; Lauretani, F.; Nouvenne, A.; Mancabelli, L.; Lugli, G. A.; Turroni, F.; Duranti, S.; Mangifesta, M.; Viappiani, A. Gut microbiota composition is associated with polypharmacy in elderly hospitalized patients. Sci. Rep. 2017, 7, 11102,  DOI: 10.1038/s41598-017-10734-y
  352. 353
    Chopra, K.; Kumar, B.; Kuhad, A. Pathobiological targets of depression. Expert Opin. Ther. Targets 2011, 15, 379400,  DOI: 10.1517/14728222.2011.553603
  353. 354
    Dinan, T. G.; Cryan, J. F. Gut-brain axis in 2016: Brain-gut-microbiota axis - mood, metabolism and behaviour. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 6970,  DOI: 10.1038/nrgastro.2016.200
  354. 355
    Foster, J. A.; McVey Neufeld, K. A. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305312,  DOI: 10.1016/j.tins.2013.01.005
  355. 356
    Halkjær, S. I.; Christensen, A. H.; Lo, B. Z. S.; Browne, P. D.; Günther, S.; Hansen, L. H.; Petersen, A. M. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut 2018, 67, 21072115,  DOI: 10.1136/gutjnl-2018-316434
  356. 357
    Holvoet, T.; Joossens, M.; Wang, J.; Boelens, J.; Verhasselt, B.; Laukens, D.; van Vlierberghe, H.; Hindryckx, P.; De Vos, M.; De Looze, D. Assessment of faecal microbial transfer in irritable bowel syndrome with severe bloating. Gut 2017, 66, 980982,  DOI: 10.1136/gutjnl-2016-312513
  357. 358
    Mizuno, S.; Masaoka, T.; Naganuma, M.; Kishimoto, T.; Kitazawa, M.; Kurokawa, S.; Nakashima, M.; Takeshita, K.; Suda, W.; Mimura, M. Bifidobacterium-Rich Fecal Donor May Be a Positive Predictor for Successful Fecal Microbiota Transplantation in Patients with Irritable Bowel Syndrome. Digestion 2017, 96, 2938,  DOI: 10.1159/000471919
  358. 359
    Zoller, V.; Laguna, A. L.; Prazeres Da Costa, O.; Buch, T.; Göke, B.; Storr, M. Fecal microbiota transfer (FMT) in a patient with refractory irritable bowel syndrome. Dtsch. Med. Wochenschr. 2015, 140, 12321236,  DOI: 10.1055/s-0041-103798
  359. 360
    Drago, L.; Meroni, G.; Pistone, D.; Pasquale, L.; Milazzo, G.; Monica, F.; Aragona, S.; Ficano, L.; Vassallo, R. Evaluation of main functional dyspepsia symptoms after probiotic administration in patients receiving conventional pharmacological therapies. J. Int. Med. Res. 2021, 49, 300060520982657,  DOI: 10.1177/0300060520982657
  360. 361
    Potter, M. D. E.; Talley, N. J. Editorial: new insights into the global prevalence of uninvestigated and functional dyspepsia. Aliment. Pharmacol. Ther. 2020, 52 (8), 14071408,  DOI: 10.1111/apt.16059
  361. 362
    Zhong, L.; Shanahan, E. R.; Raj, A.; Koloski, N. A.; Fletcher, L.; Morrison, M.; Walker, M. M.; Talley, N. J.; Holtmann, G. Dyspepsia and the microbiome: time to focus on the small intestine. Gut 2017, 66, 11681169,  DOI: 10.1136/gutjnl-2016-312574
  362. 363
    Vanheel, H.; Vicario, M.; Vanuytsel, T.; Van Oudenhove, L.; Martinez, C.; Keita, Å. V.; Pardon, N.; Santos, J.; Söderholm, J. D.; Tack, J. Impaired duodenal mucosal integrity and low-grade inflammation in functional dyspepsia. Gut 2014, 63, 262271,  DOI: 10.1136/gutjnl-2012-303857
  363. 364
    Liu, X. J.; Xie, W. R.; Wu, L. H.; Ye, Z. N.; Zhang, X. Y.; Zhang, R.; He, X. X. Changes in oral flora of patients with functional dyspepsia. Sci. Rep. 2021, 11, 8089,  DOI: 10.1038/s41598-021-87600-5
  364. 365
    Aro, P.; Talley, N. J.; Johansson, S. E.; Agréus, L.; Ronkainen, J. Anxiety Is Linked to New-Onset Dyspepsia in the Swedish Population: A 10-Year Follow-up Study. Gastroenterology 2015, 148, 928937,  DOI: 10.1053/j.gastro.2015.01.039
  365. 366
    Van Oudenhove, L.; Aziz, Q. The role of psychosocial factors and psychiatric disorders in functional dyspepsia. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 158167,  DOI: 10.1038/nrgastro.2013.10
  366. 367
    Hannibal, K. E.; Bishop, M. D. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys. Ther. 2014, 94, 18161825,  DOI: 10.2522/ptj.20130597
  367. 368
    Koloski, N. A.; Jones, M.; Kalantar, J.; Weltman, M.; Zaguirre, J.; Talley, N. J. The brain--gut pathway in functional gastrointestinal disorders is bidirectional: a 12-year prospective population-based study. Gut 2012, 61, 12841290,  DOI: 10.1136/gutjnl-2011-300474
  368. 369
    Cryan, J. F.; Dinan, T. G. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701712,  DOI: 10.1038/nrn3346
  369. 370
    Browning, K. N.; Travagli, R. A. Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. Compr Physiol 2014, 4, 13391368,  DOI: 10.1002/cphy.c130055
  370. 371
    Barberio, B.; Judge, C.; Savarino, E. V.; Ford, A. C. Global prevalence of functional constipation according to the Rome criteria: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2021, 6, 638648,  DOI: 10.1016/S2468-1253(21)00111-4
  371. 372
    Vriesman, M. H.; Koppen, I. J. N.; Camilleri, M.; Di Lorenzo, C.; Benninga, M. A. Management of functional constipation in children and adults. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 2139,  DOI: 10.1038/s41575-019-0222-y
  372. 373
    Zhuang, M.; Shang, W.; Ma, Q.; Strappe, P.; Zhou, Z. Abundance of Probiotics and Butyrate-Production Microbiome Manages Constipation via Short-Chain Fatty Acids Production and Hormones Secretion. Mol. Nutr. Food Res. 2019, 63, e1801187,  DOI: 10.1002/mnfr.201801187
  373. 374
    Attaluri, A.; Jackson, M.; Valestin, J.; Rao, S. S. Methanogenic flora is associated with altered colonic transit but not stool characteristics in constipation without IBS. Am. J. Gastroenterol. 2010, 105, 14071411,  DOI: 10.1038/ajg.2009.655
  374. 375
    Chen, Y.; Wu, T.; Lu, W.; Yuan, W.; Pan, M.; Lee, Y. K.; Zhao, J.; Zhang, H.; Chen, W.; Zhu, J. Predicting the Role of the Human Gut Microbiome in Constipation Using Machine-Learning Methods: A Meta-Analysis. Microorganisms 2021, 9, 2149,  DOI: 10.3390/microorganisms9102149
  375. 376
    Tian, H.; Ye, C.; Yang, B.; Cui, J.; Zheng, Z.; Wu, C.; Zhou, S.; Lv, X.; Qin, N.; Qin, H. Gut Metagenome as a Potential Diagnostic and Predictive Biomarker in Slow Transit Constipation. Front. Med. 2022, 8, 777961,  DOI: 10.3389/fmed.2021.777961
  376. 377
    Sartor, R. B. Optimal sampling of the intestinal microbiota for research. Nature Reviews Gastroenterology & Hepatology 2015, 12, 253254,  DOI: 10.1038/nrgastro.2015.46
  377. 378
    Swidsinski, A.; Loening-Baucke, V.; Verstraelen, H.; Osowska, S.; Doerffel, Y. Biostructure of fecal microbiota in healthy subjects and patients with chronic idiopathic diarrhea. Gastroenterology 2008, 135, 568579,  DOI: 10.1053/j.gastro.2008.04.017
  378. 379
    Durbán, A.; Abellán, J. J.; Jiménez-Hernández, N.; Salgado, P.; Ponce, M.; Ponce, J.; Garrigues, V.; Latorre, A.; Moya, A. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome. Environ. Microbiol. Rep. 2012, 4, 242247,  DOI: 10.1111/j.1758-2229.2012.00327.x
  379. 380
    Parthasarathy, G.; Chen, J.; Chen, X.; Chia, N.; O’Connor, H. M.; Wolf, P. G.; Gaskins, H. R.; Bharucha, A. E. Relationship Between Microbiota of the Colonic Mucosa vs Feces and Symptoms, Colonic Transit, and Methane Production in Female Patients With Chronic Constipation. Gastroenterology 2016, 150, 367379.e1,  DOI: 10.1053/j.gastro.2015.10.005
  380. 381
    Procházková, N.; Falony, G.; Dragsted, L. O.; Licht, T. R.; Raes, J.; Roager, H. M. Advancing human gut microbiota research by considering gut transit time. Gut 2023, 72, 180191,  DOI: 10.1136/gutjnl-2022-328166
  381. 382
    Hosseinzadeh, S. T.; Poorsaadati, S.; Radkani, B.; Forootan, M. Psychological disorders in patients with chronic constipation. Gastroenterol. Hepatol. Bed Bench 2011, 4, 159163
  382. 383
    Duan, S.; Liu, L.; Li, G.; Wang, J.; Hu, Y.; Zhang, W.; Tan, Z.; Jia, Z.; Zhang, L.; von Deneen, K. M. Altered Functional Connectivity Within and Between Salience and Sensorimotor Networks in Patients With Functional Constipation. Front. Neurosci. 2021, 15, 628880,  DOI: 10.3389/fnins.2021.628880
  383. 384
    Hu, C.; Liu, L.; Liu, L.; Zhang, J.; Hu, Y.; Zhang, W.; Ding, Y.; Wang, Y.; Zhang, Z.; von Deneen, K. M. Cortical morphometry alterations in brain regions involved in emotional, motor-control and self-referential processing in patients with functional constipation. Brain Imaging Behav. 2020, 14, 18991907,  DOI: 10.1007/s11682-019-00133-4
  384. 385
    Jin, Q.; Duan, S.; Li, G.; Sun, L.; Hu, Y.; Hu, C.; Zhao, J.; von Deneen, K. M.; Qian, L.; Wang, H. Sex-related differences in resting-state brain activity and connectivity in the orbital frontal cortex and insula in patients with functional constipation. Neurogastroenterol. Motil. 2019, 31, e13566,  DOI: 10.1111/nmo.13566
  385. 386
    Li, G.; Zhang, W.; Hu, Y.; Wang, J.; Li, J.; Jia, Z.; Zhang, L.; Sun, L.; von Deneen, K. M.; Duan, S. Distinct Basal Brain Functional Activity and Connectivity in the Emotional-Arousal Network and Thalamus in Patients With Functional Constipation Associated With Anxiety and/or Depressive Disorders. Psychosom. Med. 2021, 83, 707714,  DOI: 10.1097/PSY.0000000000000958
  386. 387
    Liu, L.; Hu, C.; Hu, Y.; Zhang, W.; Zhang, Z.; Ding, Y.; Wang, Y.; von Deneen, K. M.; Sun, L.; Wang, H. Abnormalities in the thalamo-cortical network in patients with functional constipation. Brain Imaging Behav. 2021, 15, 630642,  DOI: 10.1007/s11682-020-00273-y
  387. 388
    Peihong, M.; Tao, Y.; Zhaoxuan, H.; Sha, Y.; Li, C.; Kunnan, X.; Jingwen, C.; Likai, H.; Yuke, T.; Yuyi, G. Alterations of White Matter Network Properties in Patients With Functional Constipation. Front. Neurol. 2021, 12, 627130,  DOI: 10.3389/fneur.2021.627130
  388. 389
    Zhu, Q.; Cai, W.; Zheng, J.; Li, G.; Meng, Q.; Liu, Q.; Zhao, J.; von Deneen, K. M.; Wang, Y.; Cui, G. Distinct resting-state brain activity in patients with functional constipation. Neurosci. Lett. 2016, 632, 141146,  DOI: 10.1016/j.neulet.2016.08.042
  389. 390
    Mayer, E. A. Gut feelings: the emerging biology of gut–brain communication. Nat. Rev. Neurosci. 2011, 12, 453466,  DOI: 10.1038/nrn3071
  390. 391
    Rubio, A.; Van Oudenhove, L.; Pellissier, S.; Ly, H. G.; Dupont, P.; Lafaye de Micheaux, H.; Tack, J.; Dantzer, C.; Delon-Martin, C.; Bonaz, B. Uncertainty in anticipation of uncomfortable rectal distension is modulated by the autonomic nervous system--a fMRI study in healthy volunteers. NeuroImage 2015, 107, 1022,  DOI: 10.1016/j.neuroimage.2014.11.043
  391. 392
    Molina-Torres, G.; Rodriguez-Arrastia, M.; Roman, P.; Sanchez-Labraca, N.; Cardona, D. Stress and the gut microbiota-brain axis. Behav. Pharmacol. 2019, 30, 187200,  DOI: 10.1097/FBP.0000000000000478
  392. 393
    Vanuytsel, T.; van Wanrooy, S.; Vanheel, H.; Vanormelingen, C.; Verschueren, S.; Houben, E.; Salim Rasoel, S.; Tóth, J.; Holvoet, L.; Farré, R. Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut 2014, 63, 12931299,  DOI: 10.1136/gutjnl-2013-305690
  393. 394
    Yarandi, S. S.; Peterson, D. A.; Treisman, G. J.; Moran, T. H.; Pasricha, P. J. Modulatory Effects of Gut Microbiota on the Central Nervous System: How Gut Could Play a Role in Neuropsychiatric Health and Diseases. J. Neurogastroenterol. Motil. 2016, 22, 201212,  DOI: 10.5056/jnm15146
  394. 395
    Kelly, J. R.; Kennedy, P. J.; Cryan, J. F.; Dinan, T. G.; Clarke, G.; Hyland, N. P. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392,  DOI: 10.3389/fncel.2015.00392
  395. 396
    Tannock, G. W.; Savage, D. C. Influences of dietary and environmental stress on microbial populations in the murine gastrointestinal tract. Infect. Immun. 1974, 9, 591598,  DOI: 10.1128/iai.9.3.591-598.1974
  396. 397
    Yang, H.; Zhao, X.; Tang, S.; Huang, H.; Zhao, X.; Ning, Z.; Fu, X.; Zhang, C. Probiotics reduce psychological stress in patients before laryngeal cancer surgery. Asia Pac. J. Clin. Oncol. 2016, 12, e9296,  DOI: 10.1111/ajco.12120
  397. 398
    Karl, J. P.; Margolis, L. M.; Madslien, E. H.; Murphy, N. E.; Castellani, J. W.; Gundersen, Y.; Hoke, A. V.; Levangie, M. W.; Kumar, R.; Chakraborty, N. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G559g571,  DOI: 10.1152/ajpgi.00066.2017
  398. 399
    Kato-Kataoka, A.; Nishida, K.; Takada, M.; Kawai, M.; Kikuchi-Hayakawa, H.; Suda, K.; Ishikawa, H.; Gondo, Y.; Shimizu, K.; Matsuki, T. Fermented Milk Containing Lactobacillus casei Strain Shirota Preserves the Diversity of the Gut Microbiota and Relieves Abdominal Dysfunction in Healthy Medical Students Exposed to Academic Stress. Appl. Environ. Microbiol. 2016, 82, 36493658,  DOI: 10.1128/AEM.04134-15
  399. 400
    Takada, M.; Nishida, K.; Kataoka-Kato, A.; Gondo, Y.; Ishikawa, H.; Suda, K.; Kawai, M.; Hoshi, R.; Watanabe, O.; Igarashi, T. Probiotic Lactobacillus casei strain Shirota relieves stress-associated symptoms by modulating the gut-brain interaction in human and animal models. Neurogastroenterol. Motil. 2016, 28, 10271036,  DOI: 10.1111/nmo.12804
  400. 401
    Zijlmans, M. A.; Korpela, K.; Riksen-Walraven, J. M.; de Vos, W. M.; de Weerth, C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 2015, 53, 233245,  DOI: 10.1016/j.psyneuen.2015.01.006
  401. 402
    Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linlo̷kken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 11551162,  DOI: 10.1111/nmo.12378
  402. 403
    Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J. Altered fecal microbiota composition in patients with major depressive disorder. Brain. Behav. Immun. 2015, 48, 186194,  DOI: 10.1016/j.bbi.2015.03.016
  403. 404
    Tillisch, K.; Mayer, E. A.; Gupta, A.; Gill, Z.; Brazeilles, R.; Le Nevé, B.; van Hylckama Vlieg, J. E. T.; Guyonnet, D.; Derrien, M.; Labus, J. S. Brain Structure and Response to Emotional Stimuli as Related to Gut Microbial Profiles in Healthy Women. Psychosom. Med. 2017, 79, 905913,  DOI: 10.1097/PSY.0000000000000493
  404. 405
    Pfau, M. L.; Russo, S. J. Peripheral and Central Mechanisms of Stress Resilience. Neurobiol Stress 2015, 1, 6679,  DOI: 10.1016/j.ynstr.2014.09.004
  405. 406
    Franklin, T. B.; Saab, B. J.; Mansuy, I. M. Neural mechanisms of stress resilience and vulnerability. Neuron 2012, 75, 747761,  DOI: 10.1016/j.neuron.2012.08.016
  406. 407
    Bear, T.; Dalziel, J.; Coad, J.; Roy, N.; Butts, C.; Gopal, P. The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress. Microorganisms 2021, 9, 723,  DOI: 10.3390/microorganisms9040723
  407. 408
    Li, L. F.; Zou, H. W.; Song, B. L.; Wang, Y.; Jiang, Y.; Li, Z. L.; Niu, Q. H.; Liu, Y. J. Increased Lactobacillus Abundance Contributes to Stress Resilience in Mice Exposed to Chronic Social Defeat Stress. Neuroendocrinology 2023, 113, 563,  DOI: 10.1159/000528876
  408. 409
    Dinan, T. G.; Stanton, C.; Cryan, J. F. Psychobiotics: a novel class of psychotropic. Biol. Psychiatry 2013, 74, 720726,  DOI: 10.1016/j.biopsych.2013.05.001
  409. 410
    Chattu, V. K.; Manzar, M. D.; Kumary, S.; Burman, D.; Spence, D. W.; Pandi-Perumal, S. R. The Global Problem of Insufficient Sleep and Its Serious Public Health Implications. Healthcare 2019, 7, 1,  DOI: 10.3390/healthcare7010001
  410. 411
    Irwin, C.; McCartney, D.; Desbrow, B.; Khalesi, S. Effects of probiotics and paraprobiotics on subjective and objective sleep metrics: a systematic review and meta-analysis. Eur. J. Clin. Nutr. 2020, 74, 15361549,  DOI: 10.1038/s41430-020-0656-x
  411. 412
    Smith, R. P.; Easson, C.; Lyle, S. M.; Kapoor, R.; Donnelly, C. P.; Davidson, E. J.; Parikh, E.; Lopez, J. V.; Tartar, J. L. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019, 14, e0222394  DOI: 10.1371/journal.pone.0222394
  412. 413
    Benedict, C.; Vogel, H.; Jonas, W.; Woting, A.; Blaut, M.; Schürmann, A.; Cedernaes, J. Gut microbiota and glucometabolic alterations in response to recurrent partial sleep deprivation in normal-weight young individuals. Molecular Metabolism 2016, 5, 11751186,  DOI: 10.1016/j.molmet.2016.10.003
  413. 414
    Zhang, S. L.; Bai, L.; Goel, N.; Bailey, A.; Jang, C. J.; Bushman, F. D.; Meerlo, P.; Dinges, D. F.; Sehgal, A. Human and rat gut microbiome composition is maintained following sleep restriction. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E1564E1571,  DOI: 10.1073/pnas.1620673114
  414. 415
    Anderson, J. R.; Carroll, I.; Azcarate-Peril, M. A.; Rochette, A. D.; Heinberg, L. J.; Peat, C.; Steffen, K.; Manderino, L. M.; Mitchell, J.; Gunstad, J. A preliminary examination of gut microbiota, sleep, and cognitive flexibility in healthy older adults. Sleep Med. 2017, 38, 104107,  DOI: 10.1016/j.sleep.2017.07.018
  415. 416
    Krueger, J. M.; Opp, M. R. Sleep and Microbes. Int. Rev. Neurobiol. 2016, 131, 207225,  DOI: 10.1016/bs.irn.2016.07.003
  416. 417
    Frey, D. J.; Fleshner, M.; Wright, K. P. The effects of 40 h of total sleep deprivation on inflammatory markers in healthy young adults. Brain, Behavior, and Immunity 2007, 21, 10501057,  DOI: 10.1016/j.bbi.2007.04.003
  417. 418
    Kamada, N.; Seo, S. U.; Chen, G. Y.; Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2013, 13, 321335,  DOI: 10.1038/nri3430
  418. 419
    Yasuda, M.; Kato, S.; Yamanaka, N.; Iimori, M.; Matsumoto, K.; Utsumi, D.; Kitahara, Y.; Amagase, K.; Horie, S.; Takeuchi, K. 5-HT3 receptor antagonists ameliorate 5-fluorouracil-induced intestinal mucositis by suppression of apoptosis in murine intestinal crypt cells. Br. J. Pharmacol. 2013, 168, 13881400,  DOI: 10.1111/bph.12019
  419. 420
    Li, H.-L.; Lu, L.; Wang, X.-S.; Qin, L.-Y.; Wang, P.; Qiu, S.-P.; Wu, H.; Huang, F.; Zhang, B.-B.; Shi, H.-L. Alteration of Gut Microbiota and Inflammatory Cytokine/Chemokine Profiles in 5-Fluorouracil Induced Intestinal Mucositis. Frontiers in Cellular and Infection Microbiology 2017, 7, 455,  DOI: 10.3389/fcimb.2017.00455
  420. 421
    Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355361,  DOI: 10.1038/sj.ejcn.1602546
  421. 422
    Parkar, S. G.; Kalsbeek, A.; Cheeseman, J. F. Potential Role for the Gut Microbiota in Modulating Host Circadian Rhythms and Metabolic Health. Microorganisms 2019, 7, 41,  DOI: 10.3390/microorganisms7020041
  422. 423
    Zhao, D.; Yu, Y.; Shen, Y.; Liu, Q.; Zhao, Z.; Sharma, R.; Reiter, R. J. Melatonin Synthesis and Function: Evolutionary History in Animals and Plants. Front. Endocrinol. 2019, 10, 249,  DOI: 10.3389/fendo.2019.00249
  423. 424
    Wang, B.; Duan, R.; Duan, L. Prevalence of sleep disorder in irritable bowel syndrome: A systematic review with meta-analysis. Saudi J. Gastroenterol. 2018, 24, 141,  DOI: 10.4103/sjg.SJG_603_17
  424. 425
    Miglis, M. G. Autonomic dysfunction in primary sleep disorders. Sleep Med. 2016, 19, 4049,  DOI: 10.1016/j.sleep.2015.10.001
  425. 426
    Tobaldini, E.; Costantino, G.; Solbiati, M.; Cogliati, C.; Kara, T.; Nobili, L.; Montano, N. Sleep, sleep deprivation, autonomic nervous system and cardiovascular diseases. Neurosci Biobehav Rev. 2017, 74, 321329,  DOI: 10.1016/j.neubiorev.2016.07.004
  426. 427
    Vgontzas, A. N.; Chrousos, G. P. Sleep, the hypothalamic-pituitary-adrenal axis, and cytokines: multiple interactions and disturbances in sleep disorders. Endocrinol. Metab. Clin. North Am. 2002, 31, 1536,  DOI: 10.1016/S0889-8529(01)00005-6
  427. 428
    Schlereth, T.; Birklein, F. The sympathetic nervous system and pain. Neuromolecular Med. 2008, 10, 141147,  DOI: 10.1007/s12017-007-8018-6
  428. 429
    Wong, R. K.; Yang, C.; Song, G. H.; Wong, J.; Ho, K. Y. Melatonin regulation as a possible mechanism for probiotic (VSL#3) in irritable bowel syndrome: a randomized double-blinded placebo study. Dig. Dis. Sci. 2015, 60, 186194,  DOI: 10.1007/s10620-014-3299-8
  429. 430
    Zhao, E.; Tait, C.; Minacapelli, C. D.; Catalano, C.; Rustgi, V. K. Circadian Rhythms, the Gut Microbiome, and Metabolic Disorders. Gastro Hep Advances 2022, 1, 93105,  DOI: 10.1016/j.gastha.2021.10.008
  430. 431
    Liang, X.; Bushman, F. D.; FitzGerald, G. A. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 1047910484,  DOI: 10.1073/pnas.1501305112
  431. 432
    Paulose, J. K.; Wright, J. M.; Patel, A. G.; Cassone, V. M. Human Gut Bacteria Are Sensitive to Melatonin and Express Endogenous Circadian Rhythmicity. PLoS One 2016, 11, e0146643  DOI: 10.1371/journal.pone.0146643
  432. 433
    Mukherji, A.; Kobiita, A.; Ye, T.; Chambon, P. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell 2013, 153, 812827,  DOI: 10.1016/j.cell.2013.04.020
  433. 434
    Paschos, G. K.; FitzGerald, G. A. Circadian Clocks and Metabolism: Implications for Microbiome and Aging. Trends Genet. 2017, 33, 760769,  DOI: 10.1016/j.tig.2017.07.010
  434. 435
    Fernandez-Real, J. M.; Serino, M.; Blasco, G.; Puig, J.; Daunis-i-Estadella, J.; Ricart, W.; Burcelin, R.; Fernández-Aranda, F.; Portero-Otin, M. Gut Microbiota Interacts With Brain Microstructure and Function. J. Clin Endocrinol Metab 2015, 100, 45054513,  DOI: 10.1210/jc.2015-3076
  435. 436
    Carlson, A. L.; Xia, K.; Azcarate-Peril, M. A.; Goldman, B. D.; Ahn, M.; Styner, M. A.; Thompson, A. L.; Geng, X.; Gilmore, J. H.; Knickmeyer, R. C. Infant Gut Microbiome Associated With Cognitive Development. Biol. Psychiatry 2018, 83, 148159,  DOI: 10.1016/j.biopsych.2017.06.021
  436. 437
    Polidano, C.; Zhu, A.; Bornstein, J. C. The relation between cesarean birth and child cognitive development. Sci. Rep. 2017, 7, 11483,  DOI: 10.1038/s41598-017-10831-y
  437. 438
    Chung, Y.-C.; Jin, H.-M.; Cui, Y.; Kim, D. S.; Jung, J. M.; Park, J.-I.; Jung, E.-S.; Choi, E.-K.; Chae, S.-W. Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J. Funct. Foods 2014, 10, 465474,  DOI: 10.1016/j.jff.2014.07.007
  438. 439
    Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 13941401.e4,  DOI: 10.1053/j.gastro.2013.02.043
  439. 440
    Smith, A. P.; Sutherland, D.; Hewlett, P. An Investigation of the Acute Effects of Oligofructose-Enriched Inulin on Subjective Wellbeing, Mood and Cognitive Performance. Nutrients 2015, 7, 88878896,  DOI: 10.3390/nu7115441
  440. 441
    Lee, S.-H.; Yoon, S.-H.; Jung, Y.; Kim, N.; Min, U.; Chun, J.; Choi, I. Emotional well-being and gut microbiome profiles by enterotype. Sci. Rep. 2020, 10, 20736,  DOI: 10.1038/s41598-020-77673-z
  441. 442
    Gros, D. F.; Antony, M. M.; McCabe, R. E.; Swinson, R. P. Frequency and severity of the symptoms of irritable bowel syndrome across the anxiety disorders and depression. J. Anxiety Disord. 2009, 23, 290296,  DOI: 10.1016/j.janxdis.2008.08.004
  442. 443
    Mayer, E. A.; Labus, J. S.; Tillisch, K.; Cole, S. W.; Baldi, P. Towards a systems view of IBS. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 592605,  DOI: 10.1038/nrgastro.2015.121
  443. 444
    Zhang, J.; Ma, L.; Chang, L.; Pu, Y.; Qu, Y.; Hashimoto, K. A key role of the subdiaphragmatic vagus nerve in the depression-like phenotype and abnormal composition of gut microbiota in mice after lipopolysaccharide administration. Translational Psychiatry 2020, 10, 186,  DOI: 10.1038/s41398-020-00878-3
  444. 445
    Pu, Y.; Tan, Y.; Qu, Y.; Chang, L.; Wang, S.; Wei, Y.; Wang, X.; Hashimoto, K. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain, Behavior, and Immunity 2021, 94, 318326,  DOI: 10.1016/j.bbi.2020.12.032
  445. 446
    Bruch, J. D. Intestinal infection associated with future onset of an anxiety disorder: Results of a nationally representative study. Brain. Behav. Immun. 2016, 57, 222226,  DOI: 10.1016/j.bbi.2016.05.014
  446. 447
    Kiecolt-Glaser, J. K.; Wilson, S. J.; Shrout, M. R.; Madison, A. A.; Andridge, R.; Peng, J.; Malarkey, W. B.; Bailey, M. T. The gut reaction to couples’ relationship troubles: A route to gut dysbiosis through changes in depressive symptoms. Psychoneuroendocrinology 2021, 125, 105132,  DOI: 10.1016/j.psyneuen.2021.105132
  447. 448
    Kim, H. N.; Yun, Y.; Ryu, S.; Chang, Y.; Kwon, M. J.; Cho, J.; Shin, H.; Kim, H. L. Correlation between gut microbiota and personality in adults: A cross-sectional study. Brain. Behav. Immun. 2018, 69, 374385,  DOI: 10.1016/j.bbi.2017.12.012
  448. 449
    Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E. F.; Wang, J.; Tito, R. Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C. The neuroactive potential of the human gut microbiota in quality of life and depression. Nature Microbiology 2019, 4, 623632,  DOI: 10.1038/s41564-018-0337-x
  449. 450
    Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D. R.; Fernandes, G. R.; Tap, J.; Bruls, T.; Batto, J.-M. Enterotypes of the human gut microbiome. Nature 2011, 473, 174180,  DOI: 10.1038/nature09944
  450. 451
    Costello, E. K.; Lauber, C. L.; Hamady, M.; Fierer, N.; Gordon, J. I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 16941697,  DOI: 10.1126/science.1177486
  451. 452
    Wu, G. D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y. Y.; Keilbaugh, S. A.; Bewtra, M.; Knights, D.; Walters, W. A.; Knight, R. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105108,  DOI: 10.1126/science.1208344
  452. 453
    Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M. R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315320,  DOI: 10.1016/j.nut.2015.09.003
  453. 454
    Wallace, C. J. K.; Milev, R. The effects of probiotics on depressive symptoms in humans: a systematic review. Ann. Gen. Psychiatry 2017, 16, 14,  DOI: 10.1186/s12991-017-0138-2
  454. 455
    Le Morvan de Sequeira, C.; Hengstberger, C.; Enck, P.; Mack, I. Effect of Probiotics on Psychiatric Symptoms and Central Nervous System Functions in Human Health and Disease: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 621,  DOI: 10.3390/nu14030621
  455. 456
    Hill, C.; Guarner, F.; Reid, G.; Gibson, G. R.; Merenstein, D. J.; Pot, B.; Morelli, L.; Canani, R. B.; Flint, H. J.; Salminen, S. 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, 506514,  DOI: 10.1038/nrgastro.2014.66
  456. 457
    Dahiya, D.; Nigam, P. S. Clinical Potential of Microbial Strains, Used in Fermentation for Probiotic Food, Beverages and in Synbiotic Supplements, as Psychobiotics for Cognitive Treatment through Gut-Brain Signaling. Microorganisms 2022, 10, 1687,  DOI: 10.3390/microorganisms10091687
  457. 458
    Zawistowska-Rojek, A.; Tyski, S. How to Improve Health with Biological Agents-Narrative Review. Nutrients 2022, 14, 1700,  DOI: 10.3390/nu14091700
  458. 459
    Gibson, G. R.; Hutkins, R.; Sanders, M. E.; Prescott, S. L.; Reimer, R. A.; Salminen, S. J.; Scott, K.; Stanton, C.; Swanson, K. S.; Cani, P. D. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nature Reviews Gastroenterology & Hepatology 2017, 14, 491502,  DOI: 10.1038/nrgastro.2017.75
  459. 460
    Swanson, K. S.; Gibson, G. R.; Hutkins, R.; Reimer, R. A.; Reid, G.; Verbeke, K.; Scott, K. P.; Holscher, H. D.; Azad, M. B.; Delzenne, N. M. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nature Reviews Gastroenterology & Hepatology 2020, 17, 687701,  DOI: 10.1038/s41575-020-0344-2
  460. 461
    Salminen, S.; Collado, M. C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E. M. M.; Sanders, M. E.; Shamir, R.; Swann, J. R.; Szajewska, H. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nature Reviews Gastroenterology & Hepatology 2021, 18, 649667,  DOI: 10.1038/s41575-021-00440-6
  461. 462
    Sarkar, A.; Lehto, S. M.; Harty, S.; Dinan, T. G.; Cryan, J. F.; Burnet, P. W. J. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci. 2016, 39, 763781,  DOI: 10.1016/j.tins.2016.09.002
  462. 463
    Long-Smith, C.; O’Riordan, K. J.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Microbiota-Gut-Brain Axis: New Therapeutic Opportunities. Annu. Rev. Pharmacol Toxicol 2020, 60, 477502,  DOI: 10.1146/annurev-pharmtox-010919-023628
  463. 464
    Pferschy-Wenzig, E. M.; Pausan, M. R.; Ardjomand-Woelkart, K.; Röck, S.; Ammar, R. M.; Kelber, O.; Moissl-Eichinger, C.; Bauer, R. Medicinal Plants and Their Impact on the Gut Microbiome in Mental Health: A Systematic Review. Nutrients 2022, 14, 2111,  DOI: 10.3390/nu14102111
  464. 465
    PitchBook. https://www.pitchbook.com/ (accessed March 30, 2023).
  465. 466
    Black, C. J.; Ford, A. C. Global burden of irritable bowel syndrome: trends, predictions and risk factors. Nature Reviews Gastroenterology & Hepatology 2020, 17, 473486,  DOI: 10.1038/s41575-020-0286-8
  466. 467
    Fecal Microbiota Transplantation. https://www.idsociety.org/public-health/emerging-clinical-issues/emerging-clinical-issues/fecal-microbiota-transplantation/ (accessed November 28, 2022).
  467. 468
    BiomeBank announces world first regulatory approval for donor derived microbiome drug. https://www.biomebank.com/news/media-release/biomebank-announces-world-first-regulatory-approval-for-donor-derived-microbiome-drug/ (accessed November 22, 2022).
  468. 469
    Ferring Receives U.S. FDA Approval for REBYOTA (fecal microbiota, live-jslm) – A Novel First-in-Class Microbiota-Based Live Biotherapeutic. https://ferringusa.com/?press=ferring-receives-u-s-fda-approval-for-rebyota-fecal-microbiota-live-jslm-a-novel-first-in-class-microbiota-based-live-biotherapeutic (accessed December 6, 2022).
  469. 470
    Clinical Trials. https://www.clinicaltrials.gov/ (accessed January 27, 2023).
  470. 471
    Fijan, S. Microorganisms with claimed probiotic properties: an overview of recent literature. Int. J. Environ. Res. Public Health 2014, 11, 47454767,  DOI: 10.3390/ijerph110504745
  471. 472
    Effect of Two Probiotic Formulations on Mental Health and Mood Biomarkers in Adults With Depressive Symptoms. https://clinicaltrials.gov/ct2/show/NCT05564767?term=NCT05564767&draw=2&rank=1 (accessed November 27, 2022).
  472. 473
    Effects of Probiotics on Gut Microbiota, Endocannabinoid and Immune Activation and Symptoms of Fatigue in Dancers. https://clinicaltrials.gov/ct2/show/NCT05567653?term=NCT05567653&draw=2&rank=1 (accessed November 27, 2022).
  473. 474
    Probiotics on Sleep Among Adults Study. https://clinicaltrials.gov/ct2/show/NCT04767997?term=NCT04767997&draw=2&rank=1 (accessed November 27, 2022).
  474. 475
    Rode, J.; Edebol Carlman, H. M. T.; König, J.; Repsilber, D.; Hutchinson, A. N.; Thunberg, P.; Andersson, P.; Persson, J.; Kiselev, A.; Lathrop Stern, L. Probiotic Mixture Containing Lactobacillus helveticus, Bifidobacterium longum and Lactiplantibacillus plantarum Affects Brain Responses Toward an Emotional Task in Healthy Subjects: A Randomized Clinical Trial. Front Nutr 2022, 9, 827182,  DOI: 10.3389/fnut.2022.827182
  475. 476
    Patterson, E.; Griffin, S. M.; Ibarra, A.; Ellsiepen, E.; Hellhammer, J. Lacticaseibacillus paracasei Lpc-37® improves psychological and physiological markers of stress and anxiety in healthy adults: a randomized, double-blind, placebo-controlled and parallel clinical trial (the Sisu study). Neurobiol Stress 2020, 13, 100277,  DOI: 10.1016/j.ynstr.2020.100277
  476. 477
    Stress & Anxiety Dampening Effects of a Probiotic Supplement Compared to Placebo in Healthy Subjects. https://www.clinicaltrials.gov/ct2/show/NCT03494725?term=NCT03494725&draw=2&rank=1 (accessed November 28, 2022).
  477. 478
    The Cognitive Effects of 6 Weeks Administration With a Probiotic. https://www.clinicaltrials.gov/ct2/show/NCT03601559?term=NCT03601559&draw=2&rank=1 (accessed November 28, 2022).
  478. 479
    Probiotic Effects on the Microbe-brain-gut Interaction and Brain Activity During Stress Tasks in Healthy Subjects. https://www.clinicaltrials.gov/ct2/show/NCT03615651?term=NCT03615651&draw=2&rank=1 (accessed November 28, 2022).
  479. 480
    Lactobacillus Plantarum DR7 for Gut-Brain-Axis Benefits (DR7). https://www.clinicaltrials.gov/ct2/show/NCT03370458?term=NCT03370458&draw=2&rank=1 (accessed November 28, 2022).
  480. 481
    Sur, D.; Manna, B.; Niyogi, S. K.; Ramamurthy, T.; Palit, A.; Nomoto, K.; Takahashi, T.; Shima, T.; Tsuji, H.; Kurakawa, T. Role of probiotic in preventing acute diarrhoea in children: a community-based, randomized, double-blind placebo-controlled field trial in an urban slum. Epidemiology and Infection 2011, 139, 919926,  DOI: 10.1017/S0950268810001780
  481. 482
    Cottrell, J.; Koenig, K.; Perfekt, R.; Hofmann, R.; For the Loperamide–Simethicone Acute Diarrhoea Study, T. Comparison of Two Forms of Loperamide–Simeticone and a Probiotic Yeast (Saccharomyces boulardii) in the Treatment of Acute Diarrhoea in Adults: A Randomised Non-Inferiority Clinical Trial. Drugs R&D 2015, 15, 363373,  DOI: 10.1007/s40268-015-0111-y
  482. 483
    Skrzydło-Radomańska, B.; Prozorow-Król, B.; Cichoż-Lach, H.; Majsiak, E.; Bierła, J. B.; Kanarek, E.; Sowińska, A.; Cukrowska, B. The Effectiveness and Safety of Multi-Strain Probiotic Preparation in Patients with Diarrhea-Predominant Irritable Bowel Syndrome: A Randomized Controlled Study. Nutrients 2021, 13, 756,  DOI: 10.3390/nu13030756
  483. 484
    Quigley, E. M. M.; Markinson, L.; Stevenson, A.; Treasure, F. P.; Lacy, B. E. Randomised clinical trial: efficacy and safety of the live biotherapeutic product MRx1234 in patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 2023, 57, 8193,  DOI: 10.1111/apt.17310
  484. 485
    Effect of Lactobacillus Gasseri DSM 27123 on Functional Constipation in Healthy Women. https://www.clinicaltrials.gov/ct2/show/NCT02592200?term=NCT02592200&draw=2&rank=1 (accessed November 28, 2022).
  485. 486
    Dietary Supplementation Effects on Bowel Movement Frequency and Intestinal Biological Markers in Seniors Presenting Slowed Intestinal Transit. https://www.clinicaltrials.gov/ct2/show/NCT04304170?term=NCT04304170&draw=2&rank=1 (accessed November 28, 2022).
  486. 487
    Multi Strain Probiotic Preparation in Patients With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT04662957?term=NCT04662957&draw=2&rank=1 (accessed November 28, 2022).
  487. 488
    The Effect of Probiotic Supported Yogurt Consumption on Gastrointestinal Symptoms. https://www.clinicaltrials.gov/ct2/show/NCT05566171?term=NCT05566171&draw=2&rank=1 (accessed November 28, 2022).
  488. 489
    B. Lactis HN019 for Functional Constipation (CTT). https://www.clinicaltrials.gov/ct2/show/NCT01463293?term=NCT01463293&draw=2&rank=1 (accessed November 28, 2022).
  489. 490
    Effects of Cultura Yoghurt in Relation to Transit Time and Digestive Discomfort in Healthy Women and Men. https://www.clinicaltrials.gov/ct2/show/NCT01102036?term=NCT01102036&draw=2&rank=1 (accessed November 28, 2022).
  490. 491
    A Trial for New Treatment of Adult Participants With Irritable Bowel Syndrome. https://clinicaltrials.gov/ct2/show/NCT03721107?term=NCT03721107&draw=2&rank=1 (accessed Feb 16, 2023).
  491. 492
    Randomized Controlled Field Trial of a Probiotics to Assess Its Role in Preventig Diarrhoea (Yakult). https://www.clinicaltrials.gov/ct2/show/NCT00534170?term=NCT00534170&draw=2&rank=1 (accessed November 28, 2022).
  492. 493
    Probiotics and Hospital Outcome in the Elderly (PROAGE). https://www.clinicaltrials.gov/ct2/show/NCT00794924?term=NCT00794924&draw=2&rank=1 (accessed November 28, 2022).
  493. 494
    Evaluation of the Efficacy of Two Probiotic Strains for Irritable Bowel Syndrome (14PIHL). https://www.clinicaltrials.gov/ct2/show/NCT02213172?term=NCT02213172&draw=2&rank=1 (accessed November 28, 2022).
  494. 495
    A Comparison of Three Medications to Treat Diarrhea in Adults. https://www.clinicaltrials.gov/ct2/show/NCT00807326?term=NCT00807326&draw=2&rank=1 (accessed November 28, 2022).
  495. 496
    A Trial to Evaluate the Effects of Bifidobacterium Longum NCC3001 on Intestinal and Psychological Symptoms in Subjects With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT05054309?term=NCT05054309&draw=2&rank=1 (accessed November 28, 2022).
  496. 497
    Study to Evaluate a Probiotic in Healthy Subjects With a History of Abdominal Discomfort and Bloating. https://www.clinicaltrials.gov/ct2/show/NCT01099696?term=NCT01099696&draw=2&rank=1 (accessed November 28, 2022).
  497. 498
    Efficacy of a Multi-strain Probiotic in the Treatment of Irritable Bowel Syndrome (IBS). https://www.clinicaltrials.gov/ct2/show/NCT01887834?term=NCT01887834&draw=2&rank=1 (accessed November 28, 2022).
  498. 499
    Trial to Evaluate Dietary Supplements to Maintain Gut Health During Travel (P3). https://www.clinicaltrials.gov/ct2/show/NCT04605783?term=NCT04605783&draw=2&rank=1 (accessed November 28, 2022).
  499. 500
    To Study the Efficacy and Safety of L. Plantarum UALp-05TM in Diarrhea- Predominant-irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT04950296?term=NCT04950296&draw=2&rank=1 (accessed November 28, 2022).
  500. 501
    Saccharomyces Cerevisiae for Irritable Bowel Syndrome (IBS). https://www.clinicaltrials.gov/ct2/show/NCT05149599?term=NCT05149599&draw=2&rank=1 (accessed November 28, 2022).
  501. 502
    Johnstone, N.; Milesi, C.; Burn, O.; van den Bogert, B.; Nauta, A.; Hart, K.; Sowden, P.; Burnet, P. W. J.; Cohen Kadosh, K. Anxiolytic effects of a galacto-oligosaccharides prebiotic in healthy females (18–25 years) with corresponding changes in gut bacterial composition. Sci. Rep. 2021, 11, 8302,  DOI: 10.1038/s41598-021-87865-w
  502. 503
    Parilli-Moser, I.; Domínguez-López, I.; Trius-Soler, M.; Castellví, M.; Bosch, B.; Castro-Barquero, S.; Estruch, R.; Hurtado-Barroso, S.; Lamuela-Raventós, R. M. Consumption of peanut products improves memory and stress response in healthy adults from the ARISTOTLE study: A 6-month randomized controlled trial. Clin. Nutr. 2021, 40, 55565567,  DOI: 10.1016/j.clnu.2021.09.020
  503. 504
    Prebiotics and Stress Reduction in Women. https://www.clinicaltrials.gov/ct2/show/NCT05372601?term=NCT05372601&draw=2&rank=1 (accessed November 28, 2022).
  504. 505
    Evaluating the Effects of Prebiotics on Sleep, the Gut Microbiome, Cognition, Immune Function and Stress. https://www.clinicaltrials.gov/ct2/show/NCT05239845?term=NCT05239845&draw=2&rank=1 (accessed November 28, 2022).
  505. 506
    Healthy Prebiotic and Postbiotic Effects of Peanuts and Peanut Butter: College Intervention Trial (ARISTOTLE). https://www.clinicaltrials.gov/ct2/show/NCT04324749?term=NCT04324749&draw=2&rank=1 (accessed November 28, 2022).
  506. 507
    Polyphenols, Prebiotics, the Gut Microbiome and Stress. https://www.clinicaltrials.gov/ct2/show/NCT05528575?term=NCT05528575&draw=2&rank=1 (accessed November 28, 2022).
  507. 508
    Prebiotics and Mental Health: Behavioural. https://www.clinicaltrials.gov/ct2/show/NCT04616937?term=NCT04616937&draw=2&rank=1 (accessed November 28, 2022).
  508. 509
    Selling, J.; Swann, P.; Madsen, L. R., 2nd; Oswald, J. Improvement in Gastroesophageal Reflux Symptoms From a Food-grade Maltosyl-isomaltooligosaccharide Soluble Fiber Supplement: A Case Series. Integr. Med. 2018, 17, 4042
  509. 510
    Ansell, J.; Butts, C. A.; Paturi, G.; Eady, S. L.; Wallace, A. J.; Hedderley, D.; Gearry, R. B. Kiwifruit-derived supplements increase stool frequency in healthy adults: a randomized, double-blind, placebo-controlled study. Nutr. Res. (N.Y.) 2015, 35, 401408,  DOI: 10.1016/j.nutres.2015.04.005
  510. 511
    Blatchford, P.; Stoklosinski, H.; Eady, S.; Wallace, A.; Butts, C.; Gearry, R.; Gibson, G.; Ansell, J. Consumption of kiwifruit capsules increases Faecalibacterium prausnitzii abundance in functionally constipated individuals: a randomised controlled human trial. J. Nutr Sci. 2017, 6, e52,  DOI: 10.1017/jns.2017.52
  511. 512
    SILK, D. B. A.; DAVIS, A.; VULEVIC, J.; TZORTZIS, G.; GIBSON, G. R. Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2009, 29, 508518,  DOI: 10.1111/j.1365-2036.2008.03911.x
  512. 513
    Tolerability Study of a Novel Microbiome Therapeutic in Subjects With Gastroesophageal Reflux Disease. https://www.clinicaltrials.gov/ct2/show/NCT04491734?term=NCT04491734&draw=2&rank=1 (accessed November 28, 2022).
  513. 514
    Utility of the Administration of Chesnut and Quebracho Extract for Irritable Bowel Syndrome Diarrhea Predominant. https://www.clinicaltrials.gov/ct2/show/NCT05207618?term=NCT05207618&draw=2&rank=1 (accessed November 28, 2022).
  514. 515
    Evaluation of the Efficacy of a New Specific Infant Formula in Case of Functional Constipation. https://www.clinicaltrials.gov/ct2/show/NCT05340712?term=NCT05340712&draw=2&rank=1 (accessed November 28, 2022).
  515. 516
    ReFerm. https://referm.dk/ (accessed November 27, 2022).
  516. 517
    Effect of Postbiotic Product on Colonic Barriers in IBS. https://www.clinicaltrials.gov/ct2/show/NCT05475314?term=NCT05475314&draw=2&rank=1 (accessed November 28, 2022).
  517. 518
    Pilot Study to Assess the Effect of a Postbiotic Blend on Moderate Self-reported Anxiety (Anx). https://www.clinicaltrials.gov/ct2/show/NCT05562739?term=NCT05562739&draw=2&rank=1 (accessed November 28, 2022).
  518. 519
    Evaluating the Safety and Efficacy of the Probiotic Bifidobacterium Longum ES1 and the Post Biotic Heat-treated Bifidobacterium Longum ES1 (HT-ES1) on IBS Symptom Severity in Patients With Diarrhoea Predominant Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT05339243?term=NCT05339243&draw=2&rank=1 (accessed November 28, 2022).
  519. 520
    König, J.; Brummer, R. J. Faecal microbiota transplantation in IBS ─ new evidence for success?. Nature Reviews Gastroenterology & Hepatology 2020, 17, 199200,  DOI: 10.1038/s41575-020-0282-z
  520. 521
    El-Salhy, M.; Winkel, R.; Casen, C.; Hausken, T.; Gilja, O. H.; Hatlebakk, J. G. Efficacy of Fecal Microbiota Transplantation for Patients With Irritable Bowel Syndrome at 3 Years After Transplantation. Gastroenterology 2022, 163, 982994.e14,  DOI: 10.1053/j.gastro.2022.06.020
  521. 522
    Effects of Faecal Microbiota Transplantation in Patients With IBS. https://www.clinicaltrials.gov/ct2/show/NCT03822299?term=NCT03822299&draw=2&rank=1 (accessed November 28, 2022).
  522. 523
    Fecal Microbiota Transplantation in Patients With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT02092402?term=NCT02092402&draw=2&rank=1 (accessed November 28, 2022).
  523. 524
    RCE With FMT in the Treatment of Childhood Constipation. https://www.clinicaltrials.gov/ct2/show/NCT05035784?term=NCT05035784&draw=2&rank=1 (accessed November 28, 2022).
  524. 525
    FMT Capsules in Treatment of Patients With Insomnia Clinical Research. https://www.clinicaltrials.gov/ct2/show/NCT05427331?term=NCT05427331&draw=2&rank=1 (accessed November 28, 2022).
  525. 526
    Haridy, R. Australia gives world-first regulatory approval to fecal transplant therapy. https://newatlas.com/medical/first-approval-fecal-transplant-gut-microbiome-therapy/ (accessed November 22, 2022).
  526. 527
    Shepherd, T. A coup for poo: why the world’s first faecal transplant approval matters. The Guardian , November 12, 2022. https://www.theguardian.com/australia-news/2022/nov/13/a-coup-for-poo-why-the-worlds-first-faecal-transplant-approval-matters (accessed November 22, 2022).
  527. 528
    FDA Approves First Fecal Microbiota Product. https://www.fda.gov/news-events/press-announcements/fda-approves-first-fecal-microbiota-product (accessed December 2, 2022).
  528. 529
    Shaffer, J. P.; Nothias, L.-F.; Thompson, L. R.; Sanders, J. G.; Salido, R. A.; Couvillion, S. P.; Brejnrod, A. D.; Lejzerowicz, F.; Haiminen, N.; Huang, S. Standardized multi-omics of Earth’s microbiomes reveals microbial and metabolite diversity. Nat. Microbiol. 2022, 7, 2128,  DOI: 10.1038/s41564-022-01266-x
  529. 530
    Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R. B.-Z. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018, 174, 13881405.e21,  DOI: 10.1016/j.cell.2018.08.041
  530. 531
    Zhu, G.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. Probiotics for mild cognitive impairment and Alzheimer’s disease: a systematic review and meta-analysis. Foods 2021, 10, 1672,  DOI: 10.3390/foods10071672
  531. 532
    Białecka-Dębek, A.; Granda, D.; Szmidt, M. K.; Zielińska, D. Gut microbiota, probiotic interventions, and cognitive function in the elderly: a review of current knowledge. Nutrients 2021, 13, 2514,  DOI: 10.3390/nu13082514
  532. 533
    Munawar, N.; Ahsan, K.; Muhammad, K.; Ahmad, A.; Anwar, M. A.; Shah, I.; Al Ameri, A. K.; Al Mughairbi, F. Hidden role of gut microbiome dysbiosis in schizophrenia: Antipsychotics or psychobiotics as therapeutics?. Int. J. Mol. Sci. 2021, 22, 7671,  DOI: 10.3390/ijms22147671
  533. 534
    Cohen Kadosh, K.; Basso, M.; Knytl, P.; Johnstone, N.; Lau, J. Y. F.; Gibson, G. R. Psychobiotic interventions for anxiety in young people: a systematic review and meta-analysis, with youth consultation. Transl Psychiatry 2021, 11, 352,  DOI: 10.1038/s41398-021-01422-7
  534. 535
    Osadchiy, V.; Martin, C. R.; Mayer, E. A. The Gut-Brain Axis and the Microbiome: Mechanisms and Clinical Implications. Clin. Gastroenterol. Hepatol. 2019, 17, 322332,  DOI: 10.1016/j.cgh.2018.10.002
  535. 536
    Cussotto, S.; Strain, C. R.; Fouhy, F.; Strain, R. G.; Peterson, V. L.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Differential effects of psychotropic drugs on microbiome composition and gastrointestinal function. Psychopharmacology (Berl.) 2019, 236, 16711685,  DOI: 10.1007/s00213-018-5006-5

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 3 publications.

  1. Sergio Algar, Henar Vázquez-Villa, Pedro Aguilar-Garrido, Miguel Ángel Navarro-Aguadero, María Velasco-Estévez, Anabel Sánchez-Merino, Iván Arribas-Álvarez, Alberto Paradela, Rafael L. Giner-Arroyo, Joaquín Tamargo-Azpilicueta, Irene Díaz-Moreno, Joaquín Martínez-López, Miguel Gallardo, María L. López-Rodríguez, Bellinda Benhamú. Cancer-Stem-Cell Phenotype-Guided Discovery of a Microbiota-Inspired Synthetic Compound Targeting NPM1 for Leukemia. JACS Au 2024, Article ASAP.
  2. Rumiana Tenchov, Janet M. Sasso, Xinmei Wang, Qiongqiong Angela Zhou. Antiaging Strategies and Remedies: A Landscape of Research Progress and Promise. ACS Chemical Neuroscience 2024, 15 (3) , 408-446. https://doi.org/10.1021/acschemneuro.3c00532
  3. Rumiana Tenchov, Janet M. Sasso, Xinmei Wang, Qiongqiong Angela Zhou. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement. ACS Chemical Neuroscience 2024, 15 (1) , 1-30. https://doi.org/10.1021/acschemneuro.3c00531
  • Abstract

    Figure 1

    Figure 1. Timeline of major research and development milestones related to the microbiome. (12−30,39,49−80)

    Figure 2

    Figure 2. Journal and patent publication trends on gut microbiome research related to mental and gut health according to the CAS Content Collection. Inset: microbiome vs proteome document yearly trends.

    Figure 3

    Figure 3. Top countries publishing journal articles (A) and patents (B) related to gut microbiome research in mental and gut health.

    Figure 4

    Figure 4. Flow of patent filings related to gut microbiome research in mental and gut health from different patent assignee locations (left) to various patent offices of filing (right). The abbreviations on the right indicate the patent offices of United States (US), Australia (AU), World Intellectual Property Organization (WO), Canada (CA), Austria (AT), European Patent Office (EP), Israel (IL), Norway (NO), Hong Kong (HK), Mexico (MX), Japan (JP), Brazil (BR), Spain (ES), India (IN), Russian Federation (RU), China (CN), and Korea (KR).

    Figure 5

    Figure 5. Key concepts in the scientific publications relevant to the gut microbiome research in mental and gut health. (A) Number of publications exploring key concepts related to gut microbiome research in mental and gut health. (B) Trends in key concepts presented in the articles related to gut microbiome research in mental and gut health during the years 2016–2021. Percentages are calculated with yearly publication numbers for each key concept, normalized by the total number of publications for the same concept in the same time period.

    Figure 6

    Figure 6. Gut-microbiota-participant bacteria.

    Figure 7

    Figure 7. Representation (as number of records) of the gut bacteria phyla and species in the CAS Content Collection.

    Figure 8

    Figure 8. Distribution of the publications in the CAS Content Collection related to gut microbiome-associated diseases.

    Figure 9

    Figure 9. Trends in the number of publications concerning gut microbiome-related diseases during the years 2016–2021. Percentages are calculated with yearly publication numbers for each disease, normalized by the total number of publications for the same disease in the same time period.

    Figure 10

    Figure 10. Correlation between major classes of gut bacteria with mental and gastrointestinal disorders, as reflected in the number of associated records in the CAS Content Collection.

    Figure 11

    Figure 11. Trends in the therapeutic strategies applied for the treatment of mental and gastrointestinal disorders, as presented in the documents related to gut microbiome research during the years 2017–2021. Percentages are calculated with yearly publication numbers for each type of therapeutic intervention normalized by the total number of publications for the same intervention in the same time period.

    Figure 12

    Figure 12. Exemplary gut microbiome metabolites and their mechanism of action in gut–brain communications.

    Figure 13

    Figure 13. Gut microbiome metabolite classes and their roles in digestive system functions.

    Figure 14

    Figure 14. Top universities, research institutes, and hospitals publications (1967–2022) related to gut microbiome research in mental and gastrointestinal health: (A) journal publications and (B) patents.

    Figure 15

    Figure 15. Overall capital raised and deal counts of venture capital investment for the prebiotic, probiotic, and the microbiome field ($) (source: pitchbook.com).

    Figure 16

    Figure 16. (Left) The percentage and number of analyzed global organizations utilizing probiotics for mental disorders and DGBI treatment. (Right) The percentage and number of analyzed global organizations utilizing prebiotics for mental disorders and DGBI treatment.

    Figure 17

    Figure 17. (Left) The percentage and number of analyzed global organizations utilizing postbiotics for mental disorders and DGBI treatment. (Right) The percentage and number of analyzed global organizations utilizing fecal transplant for mental disorders and DGBI treatment.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 536 other publications.

    1. 1
      Schopf, J. W. Disparate rates, differing fates: tempo and mode of evolution changed from the Precambrian to the Phanerozoic. Proc. Natl. Acad. Sci. U. S. A. 1994, 91, 67356742,  DOI: 10.1073/pnas.91.15.6735
    2. 2
      Averill, C.; Anthony, M. A.; Baldrian, P.; Finkbeiner, F.; van den Hoogen, J.; Kiers, T.; Kohout, P.; Hirt, E.; Smith, G. R.; Crowther, T. W. Defending Earth’s terrestrial microbiome. Nat. Microbiol. 2022, 7, 1717,  DOI: 10.1038/s41564-022-01228-3
    3. 3
      O’Hara, A. M.; Shanahan, F. The gut flora as a forgotten organ. EMBO reports 2006, 7, 688693,  DOI: 10.1038/sj.embor.7400731
    4. 4
      Bäckhed, F.; Ley, R. E.; Sonnenburg, J. L.; Peterson, D. A.; Gordon, J. I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 19151920,  DOI: 10.1126/science.1104816
    5. 5
      Cénit, M. C.; Matzaraki, V.; Tigchelaar, E. F.; Zhernakova, A. Rapidly expanding knowledge on the role of the gut microbiome in health and disease. Biochim. Biophys. Acta 2014, 1842, 19811992,  DOI: 10.1016/j.bbadis.2014.05.023
    6. 6
      Vrancken, G.; Gregory, A. C.; Huys, G. R. B.; Faust, K.; Raes, J. Synthetic ecology of the human gut microbiota. Nature Reviews Microbiology 2019, 17, 754763,  DOI: 10.1038/s41579-019-0264-8
    7. 7
      Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ. 2019, 7, e7502,  DOI: 10.7717/peerj.7502
    8. 8
      Pariente, N.; York, A. Milestones in human microbiota research. https://www.nature.com/immersive/d42859-019-00041-z/index.html (accessed October 6, 2022).
    9. 9
      Malla, M. A.; Dubey, A.; Kumar, A.; Yadav, S.; Hashem, A.; Abd_Allah, E. F. Exploring the Human Microbiome: The Potential Future Role of Next-Generation Sequencing in Disease Diagnosis and Treatment. Front. Immunol. 2019, 9, 02868,  DOI: 10.3389/fimmu.2018.02868
    10. 10
      Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.-C. C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G. H. Microbiome definition re-visited: old concepts and new challenges. Microbiome 2020, 8, 103,  DOI: 10.1186/s40168-020-00875-0
    11. 11
      Butler, M. I.; Mörkl, S.; Sandhu, K. V.; Cryan, J. F.; Dinan, T. G. The Gut Microbiome and Mental Health: What Should We Tell Our Patients?: Le microbiote Intestinal et la Santé Mentale: que Devrions-Nous dire à nos Patients?. Can. J. Psychiatry. 2019, 64, 747760,  DOI: 10.1177/0706743719874168
    12. 12
      Corliss, J. O. Three Centuries of Protozoology: A Brief Tribute to its Founding Father, A. van Leeuwenhoek of Delft. Journal of Protozoology 1975, 22, 37,  DOI: 10.1111/j.1550-7408.1975.tb00934.x
    13. 13
      Leidy, J. A flora and fauna within living animals. Smithsonian Institution: WA, 1853.
    14. 14
      Savage, D. C. Microbial biota of the human intestine: a tribute to some pioneering scientists. Curr. Issues Intest. Microbiol. 2001, 2, 115
    15. 15
      Pariente, N. A field is born. 2019. https://www.nature.com/articles/d42859-019-00006-2.
    16. 16
      Micheli, P. A. Nova plantarum genera iuxta Tournefortii methodum disposita ; 1729.
    17. 17
      Drews, G. The roots of microbiology and the influence of Ferdinand Cohn on microbiology of the 19th century. FEMS Microbiol. Rev. 2000, 24, 225249,  DOI: 10.1111/j.1574-6976.2000.tb00540.x
    18. 18
      Boundless. Koch’s Postulates. https://bio.libretexts.org/Bookshelves/Microbiology/Book%3A_Microbiology_(Boundless)/10%3A_Epidemiology/10.1%3A_Principles_of_Epidemiology/10.1D%3A__Kochs_Postulates (accessed October 6, 2022).
    19. 19
      Metchnikoff, E. Prolongation of Life: Optimistic Studies ; 1923.
    20. 20
      Sonnenborn, U. Escherichia coli strain Nissle 1917─from bench to bedside and back: history of a special Escherichia coli strain with probiotic properties. FEMS Microbiol. Lett. 2016, 363, fnw212,  DOI: 10.1093/femsle/fnw212
    21. 21
      Tan, S. Y.; Tatsumura, Y. Alexander Fleming (1881–1955): Discoverer of penicillin. Singapore Med. J. 2015, 56, 366367,  DOI: 10.11622/smedj.2015105
    22. 22
      Almeida, A.; Mitchell, A. L.; Boland, M.; Forster, S. C.; Gloor, G. B.; Tarkowska, A.; Lawley, T. D.; Finn, R. D. A new genomic blueprint of the human gut microbiota. Nature 2019, 568, 499504,  DOI: 10.1038/s41586-019-0965-1
    23. 23
      Hungate, R. E., Chapter IV A Roll Tube Method for Cultivation of Strict Anaerobes. In Methods in Microbiology, Vol. 3; Norris, J. R., Ribbons, D. W., Eds.; Academic Press, 1969; pp 117132.
    24. 24
      Cat, L. A. The Decade Of The Microbiome. https://www.forbes.com/sites/linhanhcat/2019/12/31/decade-of-the-microbiome/?sh=7e621a679961 (accessed October 11, 2022).
    25. 25
      NIH Human Microbiome Project. https://www.hmpdacc.org/overview/ (accessed October 6, 2022).
    26. 26
      MetaHIT. https://www.gutmicrobiotaforhealth.com/metahit/ (accessed October 6, 2022).
    27. 27
      Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K. S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 5965,  DOI: 10.1038/nature08821
    28. 28
      Proctor, Lita M. The Human Microbiome Project in 2011 and Beyond. Cell Host Microbe 2011, 10, 287291,  DOI: 10.1016/j.chom.2011.10.001
    29. 29
      Birney, E.; Stamatoyannopoulos, J. A.; Dutta, A.; Guigó, R.; Gingeras, T. R.; Margulies, E. H.; Weng, Z.; Snyder, M.; Dermitzakis, E. T.; Stamatoyannopoulos, J. A. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799816,  DOI: 10.1038/nature05874
    30. 30
      Riesenfeld, C. S.; Schloss, P. D.; Handelsman, J. Metagenomics: Genomic Analysis of Microbial Communities. Annu. Rev. Genet. 2004, 38, 525552,  DOI: 10.1146/annurev.genet.38.072902.091216
    31. 31
      Jandhyala, S. M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 87878803,  DOI: 10.3748/wjg.v21.i29.8787
    32. 32
      Tomas, J.; Wrzosek, L.; Bouznad, N.; Bouet, S.; Mayeur, C.; Noordine, M. L.; Honvo-Houeto, E.; Langella, P.; Thomas, M.; Cherbuy, C. Primocolonization is associated with colonic epithelial maturation during conventionalization. FASEB J. 2013, 27, 645655,  DOI: 10.1096/fj.12-216861
    33. 33
      Caballero, S.; Pamer, E. G. Microbiota-mediated inflammation and antimicrobial defense in the intestine. Annu. Rev. Immunol. 2015, 33, 227256,  DOI: 10.1146/annurev-immunol-032713-120238
    34. 34
      Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Frontiers in Genetics 2015, 6, 148,  DOI: 10.3389/fgene.2015.00148
    35. 35
      Mayer, E. A.; Knight, R.; Mazmanian, S. K.; Cryan, J. F.; Tillisch, K. Gut microbes and the brain: paradigm shift in neuroscience. J. Neurosci. 2014, 34, 1549015496,  DOI: 10.1523/JNEUROSCI.3299-14.2014
    36. 36
      Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R. D.; Shanahan, F.; Dinan, T. G.; Cryan, J. F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666673,  DOI: 10.1038/mp.2012.77
    37. 37
      Bray, N. The microbiota–gut–brain axis. https://www.nature.com/articles/d42859-019-00021-3 (accessed October 6, 2022).
    38. 38
      Dinan, T. G.; Cryan, J. F. The impact of gut microbiota on brain and behaviour: implications for psychiatry. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 552558,  DOI: 10.1097/MCO.0000000000000221
    39. 39
      Bravo, J. A.; Forsythe, P.; Chew, M. V.; Escaravage, E.; Savignac, H. M.; Dinan, T. G.; Bienenstock, J.; Cryan, J. F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 1605016055,  DOI: 10.1073/pnas.1102999108
    40. 40
      Cohen-Sacks, H.; Elazar, V.; Gao, J.; Golomb, A.; Adwan, H.; Korchov, N.; Levy, R. J.; Berger, M. R.; Golomb, G. Delivery and expression of pDNA embedded in collagen matrices. J. Controlled Release 2004, 95, 309320,  DOI: 10.1016/j.jconrel.2003.11.001
    41. 41
      Rutsch, A.; Kantsjö, J. B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179,  DOI: 10.3389/fimmu.2020.604179
    42. 42
      Drossman, D. A.; Hasler, W. L. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. Gastroenterology 2016, 150, 12571261,  DOI: 10.1053/j.gastro.2016.03.035
    43. 43
      Perez-Muñoz, M. E.; Arrieta, M.-C.; Ramer-Tait, A. E.; Walter, J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017, 5, 48,  DOI: 10.1186/s40168-017-0268-4
    44. 44
      Dominguez-Bello, M. G.; Costello, E. K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 1197111975,  DOI: 10.1073/pnas.1002601107
    45. 45
      Hill, C. J.; Lynch, D. B.; Murphy, K.; Ulaszewska, M.; Jeffery, I. B.; O’Shea, C. A.; Watkins, C.; Dempsey, E.; Mattivi, F.; Tuohy, K. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome 2017, 5, 4,  DOI: 10.1186/s40168-016-0213-y
    46. 46
      Vatanen, T.; Jabbar, K. S.; Ruohtula, T.; Honkanen, J.; Avila-Pacheco, J.; Siljander, H.; Stražar, M.; Oikarinen, S.; Hyöty, H.; Ilonen, J. Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolism. Cell 2022, 185, 49214936.e4915,  DOI: 10.1016/j.cell.2022.11.023
    47. 47
      Baleato, C. L.; Ferguson, J. J. A.; Oldmeadow, C.; Mishra, G. D.; Garg, M. L. Plant-Based Dietary Patterns versus Meat Consumption and Prevalence of Impaired Glucose Intolerance and Diabetes Mellitus: A Cross-Sectional Study in Australian Women. Nutrients 2022, 14, 4152,  DOI: 10.3390/nu14194152
    48. 48
      Keaver, L.; Ruan, M.; Chen, F.; Du, M.; Ding, C.; Wang, J.; Shan, Z.; Liu, J.; Zhang, F. F. Plant- and animal-based diet quality and mortality among US adults: a cohort study. Br. J. Nutr. 2021, 125, 14051415,  DOI: 10.1017/S0007114520003670
    49. 49
      O’Mahony, S. M.; Marchesi, J. R.; Scully, P.; Codling, C.; Ceolho, A.-M.; Quigley, E. M. M.; Cryan, J. F.; Dinan, T. G. Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric Illnesses. Biol. Psychiatry 2009, 65, 263267,  DOI: 10.1016/j.biopsych.2008.06.026
    50. 50
      Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J. K. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology (Reading) 2010, 156, 32163223,  DOI: 10.1099/mic.0.040618-0
    51. 51
      Caporaso, J. G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F. D.; Costello, E. K.; Fierer, N.; Peña, A. G.; Goodrich, J. K.; Gordon, J. I. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 2010, 7, 335336,  DOI: 10.1038/nmeth.f.303
    52. 52
      Tang, L. Sequence-based identification of human-associated microbiota. https://www.nature.com/articles/d42859-019-00011-5 (accessed October 24, 2022).
    53. 53
      Collins, S. M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735742,  DOI: 10.1038/nrmicro2876
    54. 54
      Lagier, J.-C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M. Microbial culturomics: paradigm shift in the human gut microbiome study. Clinical Microbiology and Infection 2012, 18, 11851193,  DOI: 10.1111/1469-0691.12023
    55. 55
      Smith, P. M.; Howitt, M. R.; Panikov, N.; Michaud, M.; Gallini, C. A.; Bohlooly, Y. M.; Glickman, J. N.; Garrett, W. S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569573,  DOI: 10.1126/science.1241165
    56. 56
      Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232236,  DOI: 10.1038/nature12331
    57. 57
      Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J. R.; Pfeffer, K.; Coffer, P. J. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451455,  DOI: 10.1038/nature12726
    58. 58
      Donia, M. S.; Cimermancic, P.; Schulze, C. J.; Wieland Brown, L. C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R. G.; Fischbach, M. A. A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibiotics. Cell 2014, 158, 14021414,  DOI: 10.1016/j.cell.2014.08.032
    59. 59
      Lurie, I.; Yang, Y. X.; Haynes, K.; Mamtani, R.; Boursi, B. Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J. Clin. Psychiatry 2015, 76, 15221528,  DOI: 10.4088/JCP.15m09961
    60. 60
      Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786796,  DOI: 10.1038/mp.2016.44
    61. 61
      Davis, D. J.; Hecht, P. M.; Jasarevic, E.; Beversdorf, D. Q.; Will, M. J.; Fritsche, K.; Gillespie, C. H. Sex-specific effects of docosahexaenoic acid (DHA) on the microbiome and behavior of socially-isolated mice. Brain, Behavior, and Immunity 2017, 59, 3848,  DOI: 10.1016/j.bbi.2016.09.003
    62. 62
      Pasolli, E.; Asnicar, F.; Manara, S.; Zolfo, M.; Karcher, N.; Armanini, F.; Beghini, F.; Manghi, P.; Tett, A.; Ghensi, P. Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and Lifestyle. Cell 2019, 176, 649662.e20,  DOI: 10.1016/j.cell.2019.01.001
    63. 63
      Nayfach, S.; Shi, Z. J.; Seshadri, R.; Pollard, K. S.; Kyrpides, N. C. New insights from uncultivated genomes of the global human gut microbiome. Nature 2019, 568, 505510,  DOI: 10.1038/s41586-019-1058-x
    64. 64
      Pusceddu, M. M.; Barboza, M.; Schneider, M.; Stokes, P.; Sladek, J. A.; Torres-Fuentes, C.; Goldfild, L. R.; Gillis, S. E.; Brust-Mascher, I.; Rabasa, G. Nod-like receptors are critical for gut-brain axis signaling in mice. J. Physiol. 2019, 597, 57775797,  DOI: 10.1113/JP278640
    65. 65
      Pellegrini, C.; Antonioli, L.; Calderone, V.; Colucci, R.; Fornai, M.; Blandizzi, C. Microbiota-gut-brain axis in health and disease: Is NLRP3 inflammasome at the crossroads of microbiota-gut-brain communications?. Prog. Neurobiol. 2020, 191, 101806,  DOI: 10.1016/j.pneurobio.2020.101806
    66. 66
      Zhao, K.; Yao, M.; Zhang, X.; Xu, F.; Shao, X.; Wei, Y.; Wang, H. Flavonoids and intestinal microbes interact to alleviate depression. Journal of the Science of Food and Agriculture 2022, 102, 13111318,  DOI: 10.1002/jsfa.11578
    67. 67
      Segre, J. A. What does it take to satisfy Koch’s postulates two centuries later? Microbial genomics and Propionibacteria acnes. J. Invest. Dermatol. 2013, 133, 21412142,  DOI: 10.1038/jid.2013.260
    68. 68
      Eiseman, B.; Silen, W.; Bascom, G. S.; Kauvar, A. J. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958, 44, 854859
    69. 69
      Reyniers, J. A. The pure-culture concept and gnotobiotics. Ann. N.Y. Acad. Sci. 1959, 78, 316,  DOI: 10.1111/j.1749-6632.1959.tb53091.x
    70. 70
      Schaedler, R. W.; Dubos, R.; Costello, R. ASSOCIATION OF GERMFREE MICE WITH BACTERIA ISOLATED FROM NORMAL MICE. J. Exp. Med. 1965, 122, 7782,  DOI: 10.1084/jem.122.1.77
    71. 71
      Peppercorn, M. A.; Goldman, P. The Role of Intestinal Bacteria in the Metabolism of Salicylazosulfapyridine. Journal of Pharmacology and Experimental Therapeutics 1972, 181, 555562
    72. 72
      Stark, P. L.; Lee, A. The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J. Med. Microbiol. 1982, 15, 189203,  DOI: 10.1099/00222615-15-2-189
    73. 73
      Fleischmann, R. D.; Adams, M. D.; White, O.; Clayton, R. A.; Kirkness, E. F.; Kerlavage, A. R.; Bult, C. J.; Tomb, J. F.; Dougherty, B. A.; Merrick, J. M. Whole-genome random sequencing and assembly of Haemophilus influenzae Rd. Science 1995, 269, 496512,  DOI: 10.1126/science.7542800
    74. 74
      Wilson, K. H.; Blitchington, R. B. Human colonic biota studied by ribosomal DNA sequence analysis. Appl. Environ. Microbiol. 1996, 62, 22732278,  DOI: 10.1128/aem.62.7.2273-2278.1996
    75. 75
      Guarner, F.; Malagelada, J. R. Gut flora in health and disease. Lancet 2003, 361, 512519,  DOI: 10.1016/S0140-6736(03)12489-0
    76. 76
      Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229241,  DOI: 10.1016/j.cell.2004.07.002
    77. 77
      Mazmanian, S. K.; Liu, C. H.; Tzianabos, A. O.; Kasper, D. L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107118,  DOI: 10.1016/j.cell.2005.05.007
    78. 78
      Turnbaugh, P. J.; Ley, R. E.; Mahowald, M. A.; Magrini, V.; Mardis, E. R.; Gordon, J. I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 10271031,  DOI: 10.1038/nature05414
    79. 79
      Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C. P. M.; Alou, M. T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M. P. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 9197,  DOI: 10.1126/science.aan3706
    80. 80
      Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M. L.; Luke, J. J.; Gajewski, T. F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104108,  DOI: 10.1126/science.aao3290
    81. 81
      Belizário, J. E.; Napolitano, M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front. Microbiol. 2015, 6, 1050,  DOI: 10.3389/fmicb.2015.01050
    82. 82
      Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G. A. D.; Gasbarrini, A.; Mele, M. C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14,  DOI: 10.3390/microorganisms7010014
    83. 83
      Cresci, G. A. M.; Izzo, K. Chapter 4 - Gut Microbiome. In Adult Short Bowel Syndrome; Corrigan, M. L., Roberts, K., Steiger, E., Eds.; Academic Press, 2019; pp 4554.
    84. 84
      CAS Content Collection. https://www.cas.org/about/cas-content (accessed January 18, 2023).
    85. 85
      Kho, Z. Y.; Lal, S. K. The Human Gut Microbiome – A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835,  DOI: 10.3389/fmicb.2018.01835
    86. 86
      Sankar, S. A.; Lagier, J. C.; Pontarotti, P.; Raoult, D.; Fournier, P. E. The human gut microbiome, a taxonomic conundrum. Syst. Appl. Microbiol 2015, 38, 276286,  DOI: 10.1016/j.syapm.2015.03.004
    87. 87
      Manson, J. M.; Rauch, M.; Gilmore, M. S. The Commensal Microbiology of the Gastrointestinal Tract. In GI Microbiota and Regulation of the Immune System; Huffnagle, G. B., Noverr, M. C., Eds.; Landes Bioscience and Springer Science+Business Media, LLC: New York, 2008; pp 1528.
    88. 88
      Kim, D.-H. Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metab. Dispos. 2015, 43, 15811589,  DOI: 10.1124/dmd.115.063867
    89. 89
      Deng, P.; Swanson, K. S. Gut microbiota of humans, dogs and cats: current knowledge and future opportunities and challenges. Br. J. Nutr. 2015, 113 (Suppl), S617,  DOI: 10.1017/S0007114514002943
    90. 90
      Nardone, G.; Compare, D. The human gastric microbiota: Is it time to rethink the pathogenesis of stomach diseases?. United European Gastroenterol J. 2015, 3, 255260,  DOI: 10.1177/2050640614566846
    91. 91
      Ghosh, S.; Pramanik, S. Structural diversity, functional aspects and future therapeutic applications of human gut microbiome. Arch. Microbiol. 2021, 203, 52815308,  DOI: 10.1007/s00203-021-02516-y
    92. 92
      Leser, T. D.; Mo̷lbak, L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ. Microbiol. 2009, 11, 21942206,  DOI: 10.1111/j.1462-2920.2009.01941.x
    93. 93
      Linares, D. M.; Ross, P.; Stanton, C. Beneficial Microbes: The pharmacy in the gut. Bioengineered 2016, 7, 1120,  DOI: 10.1080/21655979.2015.1126015
    94. 94
      Shah, H. N.; Olsen, I.; Bernard, K.; Finegold, S. M.; Gharbia, S.; Gupta, R. S. Approaches to the study of the systematics of anaerobic, Gram-negative, non-sporeforming rods: Current status and perspectives. Anaerobe 2009, 15, 179194,  DOI: 10.1016/j.anaerobe.2009.08.003
    95. 95
      Thomas, F.; Hehemann, J.-H.; Rebuffet, E.; Czjzek, M.; Michel, G. Environmental and Gut Bacteroidetes: The Food Connection. Front. Microbiol. 2011, 2, 93,  DOI: 10.3389/fmicb.2011.00093
    96. 96
      McKee, L. S.; La Rosa, S. L.; Westereng, B.; Eijsink, V. G.; Pope, P. B.; Larsbrink, J. Polysaccharide degradation by the Bacteroidetes: mechanisms and nomenclature. Environ. Microbiol. Rep. 2021, 13, 559581,  DOI: 10.1111/1758-2229.12980
    97. 97
      Ludwig, W.; Schleifer, K.-H.; Whitman, W. B. Revised road map to the phylum Firmicutes. In Bergey’s Manual of Systematic Bacteriology: Vol. Three The Firmicutes; De Vos, P., Garrity, G. M., Jones, D., Krieg, N. R., Ludwig, W., Rainey, F. A., Schleifer, K.-H., Whitman, W. B., Eds.; Springer New York: New York, 2009; pp 113.
    98. 98
      Yang, M.; Gu, Y.; Li, L.; Liu, T.; Song, X.; Sun, Y.; Cao, X.; Wang, B.; Jiang, K.; Cao, H. Bile Acid–Gut Microbiota Axis in Inflammatory Bowel Disease: From Bench to Bedside. Nutrients 2021, 13, 3143,  DOI: 10.3390/nu13093143
    99. 99
      Barka, E. A.; Vatsa, P.; Sanchez, L.; Gaveau-Vaillant, N.; Jacquard, C.; Klenk, H.-P.; Clément, C.; Ouhdouch, Y.; Wezel, G. P. v. Taxonomy, Physiology, and Natural Products of Actinobacteria. Microbiology and Molecular Biology Reviews 2016, 80, 143,  DOI: 10.1128/MMBR.00019-15
    100. 100
      Hidalgo-Cantabrana, C.; Delgado, S.; Ruiz, L.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A. Bifidobacteria and Their Health-Promoting Effects. Microbiol Spectr 2017, 5, 5.3.21,  DOI: 10.1128/microbiolspec.BAD-0010-2016
    101. 101
      Stackebrandt, E.; Murray, R. G. E.; Trüper, H. G. Proteobacteria classis nov., a Name for the Phylogenetic Taxon That Includes the “Purple Bacteria and Their Relatives. International Journal of Systematic and Evolutionary Microbiology 1988, 38, 321325,  DOI: 10.1099/00207713-38-3-321
    102. 102
      Rizzatti, G.; Lopetuso, L. R.; Gibiino, G.; Binda, C.; Gasbarrini, A. Proteobacteria: A Common Factor in Human Diseases. BioMed. Research International 2017, 2017, 9351507,  DOI: 10.1155/2017/9351507
    103. 103
      Lee, K.-C.; Webb, R. I.; Janssen, P. H.; Sangwan, P.; Romeo, T.; Staley, J. T.; Fuerst, J. A. Phylum Verrucomicrobia representatives share a compartmentalized cell plan with members of bacterial phylum Planctomycetes. BMC Microbiol. 2009, 9, 5,  DOI: 10.1186/1471-2180-9-5
    104. 104
      Rappé, M. S.; Giovannoni, S. J. The Uncultured Microbial Majority. Annu. Rev. Microbiol. 2003, 57, 369394,  DOI: 10.1146/annurev.micro.57.030502.090759
    105. 105
      Geerlings, S. Y.; Ouwerkerk, J. P.; Koehorst, J. J.; Ritari, J.; Aalvink, S.; Stecher, B.; Schaap, P. J.; Paulin, L.; de Vos, W. M.; Belzer, C. Genomic convergence between Akkermansia muciniphila in different mammalian hosts. BMC Microbiol. 2021, 21, 298,  DOI: 10.1186/s12866-021-02360-6
    106. 106
      Liu, M.-J.; Yang, J.-Y.; Yan, Z.-H.; Hu, S.; Li, J.-Q.; Xu, Z.-X.; Jian, Y.-P. Recent findings in Akkermansia muciniphila-regulated metabolism and its role in intestinal diseases. Clin. Nutr. 2022, 41, 23332344,  DOI: 10.1016/j.clnu.2022.08.029
    107. 107
      Gupta, R. S.; Sethi, M. Phylogeny and molecular signatures for the phylum Fusobacteria and its distinct subclades. Anaerobe 2014, 28, 182198,  DOI: 10.1016/j.anaerobe.2014.06.007
    108. 108
      Madhogaria, B.; Bhowmik, P.; Kundu, A. Correlation between human gut microbiome and diseases. Infectious Medicine 2022, 1, 180191,  DOI: 10.1016/j.imj.2022.08.004
    109. 109
      Bull, M. J.; Plummer, N. T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. 2014, 13, 1722
    110. 110
      Vijay, A.; Valdes, A. M. Role of the gut microbiome in chronic diseases: a narrative review. Eur. J. Clin. Nutr. 2022, 76, 489501,  DOI: 10.1038/s41430-021-00991-6
    111. 111
      Baquero, F.; Nombela, C. The microbiome as a human organ. Clin Microbiol Infect 2012, 18 (Suppl 4), 24,  DOI: 10.1111/j.1469-0691.2012.03916.x
    112. 112
      Evans, J. M.; Morris, L. S.; Marchesi, J. R. The gut microbiome: the role of a virtual organ in the endocrinology of the host. J. Endocrinol. 2013, 218, R37R47,  DOI: 10.1530/JOE-13-0131
    113. 113
      Haseeb, A.; Shahzad, I.; Ghulam, H.; Muhammad Naeem, F.; Humaira, M.; Imtiaz, M.; Imran, M.; Saima, M.; Muhammad Irfan, U. Gut Microbiome: A New Organ System in Body. In Parasitology and Microbiology Research; Gilberto Antonio Bastidas, P., Asghar Ali, K., Eds.; IntechOpen: Rijeka, Croatia, 2019.
    114. 114
      Cani, P. D. Human gut microbiome: hopes, threats and promises. Gut 2018, 67, 17161725,  DOI: 10.1136/gutjnl-2018-316723
    115. 115
      Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A. N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T. W.; Andrews, E.; Ajami, N. J.; Bonham, K. S.; Brislawn, C. J. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655662,  DOI: 10.1038/s41586-019-1237-9
    116. 116
      Mars, R. A. T.; Yang, Y.; Ward, T.; Houtti, M.; Priya, S.; Lekatz, H. R.; Tang, X.; Sun, Z.; Kalari, K. R.; Korem, T. Longitudinal Multi-omics Reveals Subset-Specific Mechanisms Underlying Irritable Bowel Syndrome. Cell 2020, 182, 14601473.e17,  DOI: 10.1016/j.cell.2020.08.007
    117. 117
      Rinttilä, T.; Lyra, A.; Krogius-Kurikka, L.; Palva, A. Real-time PCR analysis of enteric pathogens from fecal samples of irritable bowel syndrome subjects. Gut Pathog. 2011, 3, 6,  DOI: 10.1186/1757-4749-3-6
    118. 118
      Mayer, E. A.; Nance, K.; Chen, S. The Gut-Brain Axis. Annu. Rev. Med. 2022, 73, 439453,  DOI: 10.1146/annurev-med-042320-014032
    119. 119
      Zhang, T.; Zhang, C.; Zhang, J.; Sun, F.; Duan, L. Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Front Cell Infect Microbiol 2022, 12, 859967,  DOI: 10.3389/fcimb.2022.859967
    120. 120
      Ponnusamy, K.; Choi, J. N.; Kim, J.; Lee, S. Y.; Lee, C. H. Microbial community and metabolomic comparison of irritable bowel syndrome faeces. J. Med. Microbiol. 2011, 60, 817827,  DOI: 10.1099/jmm.0.028126-0
    121. 121
      Rajilić-Stojanović, M.; Biagi, E.; Heilig, H. G.; Kajander, K.; Kekkonen, R. A.; Tims, S.; de Vos, W. M. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011, 141, 17921801,  DOI: 10.1053/j.gastro.2011.07.043
    122. 122
      Pittayanon, R.; Lau, J. T.; Yuan, Y.; Leontiadis, G. I.; Tse, F.; Surette, M.; Moayyedi, P. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. Gastroenterology 2019, 157, 97108,  DOI: 10.1053/j.gastro.2019.03.049
    123. 123
      El-Salhy, M.; Hatlebakk, J. G.; Gilja, O. H.; Bråthen Kristoffersen, A.; Hausken, T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 2020, 69, 859867,  DOI: 10.1136/gutjnl-2019-319630
    124. 124
      Wilson, B.; Rossi, M.; Kanno, T.; Parkes, G. C.; Anderson, S.; Mason, A. J.; Irving, P. M.; Lomer, M. C.; Whelan, K. β-Galactooligosaccharide in Conjunction With Low FODMAP Diet Improves Irritable Bowel Syndrome Symptoms but Reduces Fecal Bifidobacteria. Am. J. Gastroenterol. 2020, 115, 906915,  DOI: 10.14309/ajg.0000000000000641
    125. 125
      Vasant, D. H.; Paine, P. A.; Black, C. J.; Houghton, L. A.; Everitt, H. A.; Corsetti, M.; Agrawal, A.; Aziz, I.; Farmer, A. D.; Eugenicos, M. P. British Society of Gastroenterology guidelines on the management of irritable bowel syndrome. Gut 2021, 70, 12141240,  DOI: 10.1136/gutjnl-2021-324598
    126. 126
      Moayyedi, P.; Andrews, C. N.; MacQueen, G.; Korownyk, C.; Marsiglio, M.; Graff, L.; Kvern, B.; Lazarescu, A.; Liu, L.; Paterson, W. G. Canadian Association of Gastroenterology Clinical Practice Guideline for the Management of Irritable Bowel Syndrome (IBS). J. Can. Assoc Gastroenterol 2019, 2, 629,  DOI: 10.1093/jcag/gwy071
    127. 127
      Fukudo, S.; Okumura, T.; Inamori, M.; Okuyama, Y.; Kanazawa, M.; Kamiya, T.; Sato, K.; Shiotani, A.; Naito, Y.; Fujikawa, Y. Evidence-based clinical practice guidelines for irritable bowel syndrome 2020. J. Gastroenterol. 2021, 56, 193217,  DOI: 10.1007/s00535-020-01746-z
    128. 128
      Lacy, B. E.; Pimentel, M.; Brenner, D. M.; Chey, W. D.; Keefer, L. A.; Long, M. D.; Moshiree, B. ACG Clinical Guideline: Management of Irritable Bowel Syndrome. Am. J. Gastroenterol. 2021, 116, 17,  DOI: 10.14309/ajg.0000000000001036
    129. 129
      Ford, A. C.; Harris, L. A.; Lacy, B. E.; Quigley, E. M. M.; Moayyedi, P. Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2018, 48, 10441060,  DOI: 10.1111/apt.15001
    130. 130
      Caruso, R.; Lo, B. C.; Núñez, G. Host-microbiota interactions in inflammatory bowel disease. Nat. Rev. Immunol. 2020, 20, 411426,  DOI: 10.1038/s41577-019-0268-7
    131. 131
      Federici, S.; Kredo-Russo, S.; Valdés-Mas, R.; Kviatcovsky, D.; Weinstock, E.; Matiuhin, Y.; Silberberg, Y.; Atarashi, K.; Furuichi, M.; Oka, A. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell 2022, 185, 28792898.e24,  DOI: 10.1016/j.cell.2022.07.003
    132. 132
      Nagalingam, N. A.; Lynch, S. V. Role of the microbiota in inflammatory bowel diseases. Inflamm. Bowel Dis. 2012, 18, 968984,  DOI: 10.1002/ibd.21866
    133. 133
      Barcenilla, A.; Pryde, S. E.; Martin, J. C.; Duncan, S. H.; Stewart, C. S.; Henderson, C.; Flint, H. J. Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl. Environ. Microbiol. 2000, 66, 16541661,  DOI: 10.1128/AEM.66.4.1654-1661.2000
    134. 134
      Halfvarson, J.; Brislawn, C. J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W. A.; Bramer, L. M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol 2017, 2, 17004,  DOI: 10.1038/nmicrobiol.2017.4
    135. 135
      Mottawea, W.; Chiang, C.-K.; Mühlbauer, M.; Starr, A. E.; Butcher, J.; Abujamel, T.; Deeke, S. A.; Brandel, A.; Zhou, H.; Shokralla, S. Altered intestinal microbiota–host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 2016, 7, 13419,  DOI: 10.1038/ncomms13419
    136. 136
      Marchesi, J. R.; Holmes, E.; Khan, F.; Kochhar, S.; Scanlan, P.; Shanahan, F.; Wilson, I. D.; Wang, Y. Rapid and noninvasive metabonomic characterization of inflammatory bowel disease. J. Proteome Res. 2007, 6, 546551,  DOI: 10.1021/pr060470d
    137. 137
      Van Immerseel, F.; Ducatelle, R.; De Vos, M.; Boon, N.; Van De Wiele, T.; Verbeke, K.; Rutgeerts, P.; Sas, B.; Louis, P.; Flint, H. J. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. J. Med. Microbiol. 2010, 59, 141143,  DOI: 10.1099/jmm.0.017541-0
    138. 138
      Miquel, S.; Martín, R.; Rossi, O.; Bermúdez-Humarán, L. G.; Chatel, J. M.; Sokol, H.; Thomas, M.; Wells, J. M.; Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013, 16, 255261,  DOI: 10.1016/j.mib.2013.06.003
    139. 139
      Tamanai-Shacoori, Z.; Smida, I.; Bousarghin, L.; Loreal, O.; Meuric, V.; Fong, S. B.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Roseburia spp.: a marker of health?. Future Microbiol. 2017, 12, 157170,  DOI: 10.2217/fmb-2016-0130
    140. 140
      Li, Y.; Xia, S.; Jiang, X.; Feng, C.; Gong, S.; Ma, J.; Fang, Z.; Yin, J.; Yin, Y. Gut Microbiota and Diarrhea: An Updated Review. Microbiology 2021, 11, 625210,  DOI: 10.3389/fcimb.2021.625210
    141. 141
      Hodges, K.; Gill, R. Infectious diarrhea: Cellular and molecular mechanisms. Gut Microbes 2010, 1, 421,  DOI: 10.4161/gmic.1.1.11036
    142. 142
      Levine, M. M. Escherichia coli that cause diarrhea: enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic, and enteroadherent. J. Infect. Dis. 1987, 155, 377389,  DOI: 10.1093/infdis/155.3.377
    143. 143
      Gallo, A.; Passaro, G.; Gasbarrini, A.; Landolfi, R.; Montalto, M. Modulation of microbiota as treatment for intestinal inflammatory disorders: An uptodate. World J. Gastroenterol. 2016, 22, 71867202,  DOI: 10.3748/wjg.v22.i32.7186
    144. 144
      Bron, P. A.; Kleerebezem, M.; Brummer, R. J.; Cani, P. D.; Mercenier, A.; MacDonald, T. T.; Garcia-Ródenas, C. L.; Wells, J. M. Can probiotics modulate human disease by impacting intestinal barrier function?. Br. J. Nutr. 2017, 117, 93107,  DOI: 10.1017/S0007114516004037
    145. 145
      Zhang, S.; Wang, R.; Li, D.; Zhao, L.; Zhu, L. Role of gut microbiota in functional constipation. Gastroenterol. Rep. 2021, 9, 392401,  DOI: 10.1093/gastro/goab035
    146. 146
      Zoppi, G.; Cinquetti, M.; Luciano, A.; Benini, A.; Muner, A.; Bertazzoni Minelli, E. The intestinal ecosystem in chronic functional constipation. Acta Paediatr. 1998, 87, 836841,  DOI: 10.1111/j.1651-2227.1998.tb01547.x
    147. 147
      Khalif, I. L.; Quigley, E. M.; Konovitch, E. A.; Maximova, I. D. Alterations in the colonic flora and intestinal permeability and evidence of immune activation in chronic constipation. Dig. Liver Dis. 2005, 37, 838849,  DOI: 10.1016/j.dld.2005.06.008
    148. 148
      Kim, S. E.; Choi, S. C.; Park, K. S.; Park, M. I.; Shin, J. E.; Lee, T. H.; Jung, K. W.; Koo, H. S.; Myung, S. J. Change of Fecal Flora and Effectiveness of the Short-term VSL#3 Probiotic Treatment in Patients With Functional Constipation. J. Neurogastroenterol. Motil. 2015, 21, 111120,  DOI: 10.5056/jnm14048
    149. 149
      Zhu, L.; Liu, W.; Alkhouri, R.; Baker, R. D.; Bard, J. E.; Quigley, E. M.; Baker, S. S. Structural changes in the gut microbiome of constipated patients. Physiol. Genomics 2014, 46, 679686,  DOI: 10.1152/physiolgenomics.00082.2014
    150. 150
      Mancabelli, L.; Milani, C.; Lugli, G. A.; Turroni, F.; Mangifesta, M.; Viappiani, A.; Ticinesi, A.; Nouvenne, A.; Meschi, T.; van Sinderen, D. Unveiling the gut microbiota composition and functionality associated with constipation through metagenomic analyses. Sci. Rep. 2017, 7, 9879,  DOI: 10.1038/s41598-017-10663-w
    151. 151
      Naseer, M.; Poola, S.; Uraz, S.; Tahan, V. Therapeutic Effects of Prebiotics on Constipation: A Schematic Review. Curr. Clin. Pharmacol. 2020, 15, 207215,  DOI: 10.2174/1574884715666200212125035
    152. 152
      Zhao, Y.; Yu, Y. B. Intestinal microbiota and chronic constipation. SpringerPlus 2016, 5, 1130,  DOI: 10.1186/s40064-016-2821-1
    153. 153
      Wang, J. K.; Yao, S. K. Roles of Gut Microbiota and Metabolites in Pathogenesis of Functional Constipation. Evid. Based Complement. Alternat. Med. 2021, 2021, 5560310,  DOI: 10.1155/2021/5560310
    154. 154
      Rao, S. S.; Rattanakovit, K.; Patcharatrakul, T. Diagnosis and management of chronic constipation in adults. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 295305,  DOI: 10.1038/nrgastro.2016.53
    155. 155
      Moloney, R. D.; Desbonnet, L.; Clarke, G.; Dinan, T. G.; Cryan, J. F. The microbiome: stress, health and disease. Mamm. Genome 2014, 25, 4974,  DOI: 10.1007/s00335-013-9488-5
    156. 156
      Mayer, E. A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Invest. 2015, 125, 926938,  DOI: 10.1172/JCI76304
    157. 157
      Maldonado-Contreras, A.; Noel, S. E.; Ward, D. V.; Velez, M.; Mangano, K. M. Associations between Diet, the Gut Microbiome, and Short-Chain Fatty Acid Production among Older Caribbean Latino Adults. J. Acad. Nutr. Diet. 2020, 120, 20472060.e46,  DOI: 10.1016/j.jand.2020.04.018
    158. 158
      Zhang, H.; Chen, Y.; Wang, Z.; Xie, G.; Liu, M.; Yuan, B.; Chai, H.; Wang, W.; Cheng, P. Implications of Gut Microbiota in Neurodegenerative Diseases. Front. Immunol. 2022, 13, 785644,  DOI: 10.3389/fimmu.2022.785644
    159. 159
      Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98104,  DOI: 10.1016/j.jneuroim.2019.01.004
    160. 160
      Caspani, G.; Kennedy, S.; Foster, J. A.; Swann, J. Gut microbial metabolites in depression: understanding the biochemical mechanisms. Microbial cell (Graz, Austria) 2019, 6, 454481,  DOI: 10.15698/mic2019.10.693
    161. 161
      Roubalová, R.; Procházková, P.; Papežová, H.; Smitka, K.; Bilej, M.; Tlaskalová-Hogenová, H. Anorexia nervosa: Gut microbiota-immune-brain interactions. Clin. Nutr. 2020, 39, 676684,  DOI: 10.1016/j.clnu.2019.03.023
    162. 162
      Zhang, L.; Wang, Y.; Xiayu, X.; Shi, C.; Chen, W.; Song, N.; Fu, X.; Zhou, R.; Xu, Y. F.; Huang, L. Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2017, 60, 12411257,  DOI: 10.3233/JAD-170020
    163. 163
      Wang, L.; Christophersen, C. T.; Sorich, M. J.; Gerber, J. P.; Angley, M. T.; Conlon, M. A. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig. Dis. Sci. 2012, 57, 20962102,  DOI: 10.1007/s10620-012-2167-7
    164. 164
      Galland, L. The gut microbiome and the brain. J. Med. Food 2014, 17, 12611272,  DOI: 10.1089/jmf.2014.7000
    165. 165
      Heneka, M. T.; Carson, M. J.; El Khoury, J.; Landreth, G. E.; Brosseron, F.; Feinstein, D. L.; Jacobs, A. H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R. M. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388405,  DOI: 10.1016/S1474-4422(15)70016-5
    166. 166
      Ransohoff, R. M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777783,  DOI: 10.1126/science.aag2590
    167. 167
      Karlsson, O.; Roman, E.; Berg, A. L.; Brittebo, E. B. Early hippocampal cell death, and late learning and memory deficits in rats exposed to the environmental toxin BMAA (β-N-methylamino-L-alanine) during the neonatal period. Behav. Brain Res. 2011, 219, 310320,  DOI: 10.1016/j.bbr.2011.01.056
    168. 168
      Nunes-Costa, D.; Magalhães, J. D.; G-Fernandes, M.; Cardoso, S. M.; Empadinhas, N. Microbial BMAA and the Pathway for Parkinson’s Disease Neurodegeneration. Front. Aging Neurosci. 2020, 12, 26,  DOI: 10.3389/fnagi.2020.00026
    169. 169
      Braak, H.; Rüb, U.; Gai, W. P.; Del Tredici, K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm (Vienna) 2003, 110, 517536,  DOI: 10.1007/s00702-002-0808-2
    170. 170
      Wallen, Z. D.; Demirkan, A.; Twa, G.; Cohen, G.; Dean, M. N.; Standaert, D. G.; Sampson, T. R.; Payami, H. Metagenomics of Parkinson’s disease implicates the gut microbiome in multiple disease mechanisms. Nat. Commun. 2022, 13, 6958,  DOI: 10.1038/s41467-022-34667-x
    171. 171
      Vogt, N. M.; Kerby, R. L.; Dill-McFarland, K. A.; Harding, S. J.; Merluzzi, A. P.; Johnson, S. C.; Carlsson, C. M.; Asthana, S.; Zetterberg, H.; Blennow, K. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537,  DOI: 10.1038/s41598-017-13601-y
    172. 172
      Zhuang, Z. Q.; Shen, L. L.; Li, W. W.; Fu, X.; Zeng, F.; Gui, L.; Lü, Y.; Cai, M.; Zhu, C.; Tan, Y. L. Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J. Alzheimers Dis. 2018, 63, 13371346,  DOI: 10.3233/JAD-180176
    173. 173
      Manderino, L.; Carroll, I.; Azcarate-Peril, M. A.; Rochette, A.; Heinberg, L.; Peat, C.; Steffen, K.; Mitchell, J.; Gunstad, J. Preliminary Evidence for an Association Between the Composition of the Gut Microbiome and Cognitive Function in Neurologically Healthy Older Adults. J. Int. Neuropsychol. Soc. 2017, 23, 700705,  DOI: 10.1017/S1355617717000492
    174. 174
      Barbosa, R. S. D.; Vieira-Coelho, M. A. Probiotics and prebiotics: focus on psychiatric disorders - a systematic review. Nutr. Rev. 2020, 78, 437450,  DOI: 10.1093/nutrit/nuz080
    175. 175
      Ansari, F.; Pourjafar, H.; Tabrizi, A.; Homayouni, A. The Effects of Probiotics and Prebiotics on Mental Disorders: A Review on Depression, Anxiety, Alzheimer, and Autism Spectrum Disorders. Curr. Pharm. Biotechnol. 2020, 21, 555565,  DOI: 10.2174/1389201021666200107113812
    176. 176
      Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J. F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755764,  DOI: 10.1017/S0007114510004319
    177. 177
      Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; Bahmani, F.; Kouchaki, E.; Tamtaji, O. R.; Hamidi, G. A.; Salami, M. Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2016, 8, 256,  DOI: 10.3389/fnagi.2016.00256
    178. 178
      Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, Kc01kc05,  DOI: 10.7860/JCDR/2017/26106.10428
    179. 179
      Tamtaji, O. R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 10311035,  DOI: 10.1016/j.clnu.2018.05.018
    180. 180
      Frazier, K.; Chang, E. B. Intersection of the Gut Microbiome and Circadian Rhythms in Metabolism. Trends Endocrinol Metab 2020, 31, 2536,  DOI: 10.1016/j.tem.2019.08.013
    181. 181
      Gutierrez Lopez, D. E.; Lashinger, L. M.; Weinstock, G. M.; Bray, M. S. Circadian rhythms and the gut microbiome synchronize the host’s metabolic response to diet. Cell Metab. 2021, 33, 873887,  DOI: 10.1016/j.cmet.2021.03.015
    182. 182
      Leng, Y.; Musiek, E. S.; Hu, K.; Cappuccio, F. P.; Yaffe, K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol. 2019, 18, 307318,  DOI: 10.1016/S1474-4422(18)30461-7
    183. 183
      Leone, V.; Gibbons, S. M.; Martinez, K.; Hutchison, A. L.; Huang, E. Y.; Cham, C. M.; Pierre, J. F.; Heneghan, A. F.; Nadimpalli, A.; Hubert, N. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe 2015, 17, 681689,  DOI: 10.1016/j.chom.2015.03.006
    184. 184
      Nicholson, J. K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 12621267,  DOI: 10.1126/science.1223813
    185. 185
      Moran, C. P.; Shanahan, F. Gut microbiota and obesity: role in aetiology and potential therapeutic target. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 585597,  DOI: 10.1016/j.bpg.2014.07.005
    186. 186
      Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 5560,  DOI: 10.1038/nature11450
    187. 187
      Donohoe, D. R.; Wali, A.; Brylawski, B. P.; Bultman, S. J. Microbial regulation of glucose metabolism and cell-cycle progression in mammalian colonocytes. PLoS One 2012, 7, e46589,  DOI: 10.1371/journal.pone.0046589
    188. 188
      Donohoe, D. R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T. M.; Bunger, M. K.; Bultman, S. J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517526,  DOI: 10.1016/j.cmet.2011.02.018
    189. 189
      Tolhurst, G.; Heffron, H.; Lam, Y. S.; Parker, H. E.; Habib, A. M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F. M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364371,  DOI: 10.2337/db11-1019
    190. 190
      de Vos, W. M.; Tilg, H.; Van Hul, M.; Cani, P. D. Gut microbiome and health: mechanistic insights. Gut 2022, 71, 10201032,  DOI: 10.1136/gutjnl-2021-326789
    191. 191
      Yeoh, Y. K.; Zuo, T.; Lui, G. C.-Y.; Zhang, F.; Liu, Q.; Li, A. Y.; Chung, A. C.; Cheung, C. P.; Tso, E. Y.; Fung, K. S. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698706,  DOI: 10.1136/gutjnl-2020-323020
    192. 192
      Burchill, E.; Lymberopoulos, E.; Menozzi, E.; Budhdeo, S.; McIlroy, J. R.; Macnaughtan, J.; Sharma, N. The Unique Impact of COVID-19 on Human Gut Microbiome Research. Frontiers in Medicine 2021, 8, 652464,  DOI: 10.3389/fmed.2021.652464
    193. 193
      Bernard-Raichon, L.; Venzon, M.; Klein, J.; Axelrad, J. E.; Zhang, C.; Sullivan, A. P.; Hussey, G. A.; Casanovas-Massana, A.; Noval, M. G.; Valero-Jimenez, A. M. Gut microbiome dysbiosis in antibiotic-treated COVID-19 patients is associated with microbial translocation and bacteremia. Nat. Commun. 2022, 13, 5926,  DOI: 10.1038/s41467-022-33395-6
    194. 194
      Chumpitazi, B. P. The gut microbiome as a predictor of low fermentable oligosaccharides disaccharides monosaccharides and polyols diet efficacy in functional bowel disorders. Curr. Opin. Gastroenterol. 2020, 36, 147154,  DOI: 10.1097/MOG.0000000000000608
    195. 195
      Leshem, A.; Segal, E.; Elinav, E. The Gut Microbiome and Individual-Specific Responses to Diet. mSystems 2020, 5, e0066500620,  DOI: 10.1128/mSystems.00665-20
    196. 196
      Lepage, P.; Häsler, R.; Spehlmann, M. E.; Rehman, A.; Zvirbliene, A.; Begun, A.; Ott, S.; Kupcinskas, L.; Doré, J.; Raedler, A. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 2011, 141, 227236,  DOI: 10.1053/j.gastro.2011.04.011
    197. 197
      Manichanh, C.; Borruel, N.; Casellas, F.; Guarner, F. The gut microbiota in IBD. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 599608,  DOI: 10.1038/nrgastro.2012.152
    198. 198
      Tong, M.; Li, X.; Wegener Parfrey, L.; Roth, B.; Ippoliti, A.; Wei, B.; Borneman, J.; McGovern, D. P.; Frank, D. N.; Li, E. A modular organization of the human intestinal mucosal microbiota and its association with inflammatory bowel disease. PLoS One 2013, 8, e80702,  DOI: 10.1371/journal.pone.0080702
    199. 199
      Guinane, C. M.; Cotter, P. D. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap. Adv. Gastroenterol. 2013, 6, 295308,  DOI: 10.1177/1756283X13482996
    200. 200
      Kassam, Z.; Lee, C. H.; Yuan, Y.; Hunt, R. H. Fecal microbiota transplantation for Clostridium difficile infection: systematic review and meta-analysis. Am. J. Gastroenterol. 2013, 108, 500508,  DOI: 10.1038/ajg.2013.59
    201. 201
      Tang, W. H.; Kitai, T.; Hazen, S. L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 11831196,  DOI: 10.1161/CIRCRESAHA.117.309715
    202. 202
      Jiang, H. Y.; Zhang, X.; Yu, Z. H.; Zhang, Z.; Deng, M.; Zhao, J. H.; Ruan, B. Altered gut microbiota profile in patients with generalized anxiety disorder. J. Psychiatr. Res. 2018, 104, 130136,  DOI: 10.1016/j.jpsychires.2018.07.007
    203. 203
      Chen, Y. H.; Bai, J.; Wu, D.; Yu, S. F.; Qiang, X. L.; Bai, H.; Wang, H. N.; Peng, Z. W. Association between fecal microbiota and generalized anxiety disorder: Severity and early treatment response. J. Affect. Disord. 2019, 259, 5666,  DOI: 10.1016/j.jad.2019.08.014
    204. 204
      Hemmings, S. M. J.; Malan-Müller, S.; van den Heuvel, L. L.; Demmitt, B. A.; Stanislawski, M. A.; Smith, D. G.; Bohr, A. D.; Stamper, C. E.; Hyde, E. R.; Morton, J. T. The Microbiome in Posttraumatic Stress Disorder and Trauma-Exposed Controls: An Exploratory Study. Psychosom. Med. 2017, 79, 936946,  DOI: 10.1097/PSY.0000000000000512
    205. 205
      Stevens, B. R.; Goel, R.; Seungbum, K.; Richards, E. M.; Holbert, R. C.; Pepine, C. J.; Raizada, M. K. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut 2018, 67, 15551557,  DOI: 10.1136/gutjnl-2017-314759
    206. 206
      Kelly, J. R.; Borre, Y.; O'Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P. J.; Beers, S.; Scott, K.; Moloney, G. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109118,  DOI: 10.1016/j.jpsychires.2016.07.019
    207. 207
      Lin, P.; Ding, B.; Feng, C.; Yin, S.; Zhang, T.; Qi, X.; Lv, H.; Guo, X.; Dong, K.; Zhu, Y. Prevotella and Klebsiella proportions in fecal microbial communities are potential characteristic parameters for patients with major depressive disorder. J. Affect. Disord. 2017, 207, 300304,  DOI: 10.1016/j.jad.2016.09.051
    208. 208
      Gough, E.; Shaikh, H.; Manges, A. R. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin. Infect. Dis. 2011, 53, 9941002,  DOI: 10.1093/cid/cir632
    209. 209
      Craciun, C. I.; Neag, M. A.; Catinean, A.; Mitre, A. O.; Rusu, A.; Bala, C.; Roman, G.; Buzoianu, A. D.; Muntean, D. M.; Craciun, A. E. The Relationships between Gut Microbiota and Diabetes Mellitus, and Treatments for Diabetes Mellitus. Biomedicines 2022, 10, 308,  DOI: 10.3390/biomedicines10020308
    210. 210
      van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E. G.; de Vos, W. M.; Visser, C. E.; Kuijper, E. J.; Bartelsman, J. F. W. M.; Tijssen, J. G. P. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. New England Journal of Medicine 2013, 368, 407415,  DOI: 10.1056/NEJMoa1205037
    211. 211
      Hamilton, M. J.; Weingarden, A. R.; Sadowsky, M. J.; Khoruts, A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 761767,  DOI: 10.1038/ajg.2011.482
    212. 212
      Zafar, H.; Saier, M. H., Jr. Gut Bacteroides species in health and disease. Gut Microbes 2021, 13, 1848158,  DOI: 10.1080/19490976.2020.1848158
    213. 213
      Chen, Y.; Yang, Y.; Gu, J. Clinical Implications of the Associations Between Intestinal Microbiome and Colorectal Cancer Progression. Cancer Manag. Res. 2020, 12, 41174128,  DOI: 10.2147/CMAR.S240108
    214. 214
      Bullman, S.; Pedamallu, C. S.; Sicinska, E.; Clancy, T. E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 14431448,  DOI: 10.1126/science.aal5240
    215. 215
      Cammann, D.; Lu, Y.; Cummings, M. J.; Zhang, M. L.; Cue, J. M.; Do, J.; Ebersole, J.; Chen, X.; Oh, E. C.; Cummings, J. L. Genetic correlations between Alzheimer’s disease and gut microbiome genera. Sci. Rep. 2023, 13, 5258,  DOI: 10.1038/s41598-023-31730-5
    216. 216
      Silva, Y. P.; Bernardi, A.; Frozza, R. L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25,  DOI: 10.3389/fendo.2020.00025
    217. 217
      Manning, L. P.; Yao, C. K.; Biesiekierski, J. R. Therapy of IBS: Is a Low FODMAP Diet the Answer?. Front. Psychiatry 2020, 11, 865,  DOI: 10.3389/fpsyt.2020.00865
    218. 218
      Nanayakkara, W. S.; Skidmore, P. M.; O’Brien, L.; Wilkinson, T. J.; Gearry, R. B. Efficacy of the low FODMAP diet for treating irritable bowel syndrome: the evidence to date. Clin. Exp. Gastroenterol. 2016, 9, 131142,  DOI: 10.2147/CEG.S86798
    219. 219
      Tegegne, B. A.; Kebede, B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022, 8, e09725  DOI: 10.1016/j.heliyon.2022.e09725
    220. 220
      Aponte, M.; Murru, N.; Shoukat, M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front. Microbiol. 2020, 11, 562048,  DOI: 10.3389/fmicb.2020.562048
    221. 221
      Noonan, S.; Zaveri, M.; Macaninch, E.; Martyn, K. Food & mood: a review of supplementary prebiotic and probiotic interventions in the treatment of anxiety and depression in adults. BMJ. Nutrition, Prevention & Health 2020, 3, 351,  DOI: 10.1136/bmjnph-2019-000053
    222. 222
      MACFARLANE, S.; MACFARLANE, G. T.; CUMMINGS, J. H. Review article: prebiotics in the gastrointestinal tract. Aliment. Pharmacol. Ther. 2006, 24, 701714,  DOI: 10.1111/j.1365-2036.2006.03042.x
    223. 223
      Dinan, K.; Dinan, T. Antibiotics and mental health: The good, the bad and the ugly. J. Int. Med. 2022, 292, 858869,  DOI: 10.1111/joim.13543
    224. 224
      Karakan, T.; Ozkul, C.; Küpeli Akkol, E.; Bilici, S.; Sobarzo-Sánchez, E.; Capasso, R. Gut-Brain-Microbiota Axis: Antibiotics and Functional Gastrointestinal Disorders. Nutrients 2021, 13, 389,  DOI: 10.3390/nu13020389
    225. 225
      Xu, H. M.; Huang, H. L.; Zhou, Y. L.; Zhao, H. L.; Xu, J.; Shou, D. W.; Liu, Y. D.; Zhou, Y. J.; Nie, Y. Q. Fecal Microbiota Transplantation: A New Therapeutic Attempt from the Gut to the Brain. Gastroenterol. Res. Pract. 2021, 2021, 6699268,  DOI: 10.1155/2021/6699268
    226. 226
      Doll, J. P. K.; Vázquez-Castellanos, J. F.; Schaub, A.-C.; Schweinfurth, N.; Kettelhack, C.; Schneider, E.; Yamanbaeva, G.; Mählmann, L.; Brand, S.; Beglinger, C. Fecal Microbiota Transplantation (FMT) as an Adjunctive Therapy for Depression─Case Report. Front. Psychiatry 2022, 13, 815422,  DOI: 10.3389/fpsyt.2022.815422
    227. 227
      Waller, K. M. J.; Leong, R. W.; Paramsothy, S. An update on fecal microbiota transplantation for the treatment of gastrointestinal diseases. J. Gastroenterol. Hepatol. 2022, 37, 246255,  DOI: 10.1111/jgh.15731
    228. 228
      Hetterich, L.; Stengel, A. Psychotherapeutic Interventions in Irritable Bowel Syndrome. Front. Psychiatry 2020, 11, 286,  DOI: 10.3389/fpsyt.2020.00286
    229. 229
      Wildes, J. E.; Bedell, A.; Graham, A. K.; Kells, M. Brain-gut psychotherapies: Promising tools to address gastrointestinal problems in patients with eating disorders. Int. J. Eat. Disord. 2021, 54, 10631067,  DOI: 10.1002/eat.23555
    230. 230
      Laird, K. T.; Tanner-Smith, E. E.; Russell, A. C.; Hollon, S. D.; Walker, L. S. Comparative efficacy of psychological therapies for improving mental health and daily functioning in irritable bowel syndrome: A systematic review and meta-analysis. Clin. Psychol. Rev. 2017, 51, 142152,  DOI: 10.1016/j.cpr.2016.11.001
    231. 231
      Colomier, E.; Algera, J.; Melchior, C. Pharmacological Therapies and Their Clinical Targets in Irritable Bowel Syndrome With Diarrhea. Front. Pharmacol. 2021, 11, 629026,  DOI: 10.3389/fphar.2020.629026
    232. 232
      World Health Organization. Pharmacological treatment of mental disorders in primary health care. WHO Press: Geneva, Switzerland, 2009.
    233. 233
      Fikree, A.; Byrne, P. Management of functional gastrointestinal disorders. Clin Med. (Lond) 2021, 21, 4452,  DOI: 10.7861/clinmed.2020-0980
    234. 234
      Tierney, B. T.; Yang, Z.; Luber, J. M.; Beaudin, M.; Wibowo, M. C.; Baek, C.; Mehlenbacher, E.; Patel, C. J.; Kostic, A. D. The Landscape of Genetic Content in the Gut and Oral Human Microbiome. Cell Host Microbe 2019, 26, 283295.e288,  DOI: 10.1016/j.chom.2019.07.008
    235. 235
      International Human Genome Sequencing Consortium Finishing the euchromatic sequence of the human genome. Nature 2004, 431, 931945,  DOI: 10.1038/nature03001
    236. 236
      Liu, J.; Tan, Y.; Cheng, H.; Zhang, D.; Feng, W.; Peng, C. Functions of Gut Microbiota Metabolites, Current Status and Future Perspectives. Aging Dis. 2022, 13, 11061126,  DOI: 10.14336/AD.2022.0104
    237. 237
      Heijtz, R. D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M. L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 30473052,  DOI: 10.1073/pnas.1010529108
    238. 238
      Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.-N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. Journal of Physiology 2004, 558, 263275,  DOI: 10.1113/jphysiol.2004.063388
    239. 239
      Needham, B. D.; Funabashi, M.; Adame, M. D.; Wang, Z.; Boktor, J. C.; Haney, J.; Wu, W.-L.; Rabut, C.; Ladinsky, M. S.; Hwang, S.-J. A gut-derived metabolite alters brain activity and anxiety behaviour in mice. Nature 2022, 602, 647653,  DOI: 10.1038/s41586-022-04396-8
    240. 240
      Gacias, M.; Gaspari, S.; Santos, P.-M. G.; Tamburini, S.; Andrade, M.; Zhang, F.; Shen, N.; Tolstikov, V.; Kiebish, M. A.; Dupree, J. L. Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior. eLife 2016, 5, e13442,  DOI: 10.7554/eLife.13442
    241. 241
      Vuong, H. E.; Pronovost, G. N.; Williams, D. W.; Coley, E. J. L.; Siegler, E. L.; Qiu, A.; Kazantsev, M.; Wilson, C. J.; Rendon, T.; Hsiao, E. Y. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020, 586, 281286,  DOI: 10.1038/s41586-020-2745-3
    242. 242
      Wei, G. Z.; Martin, K. A.; Xing, P. Y.; Agrawal, R.; Whiley, L.; Wood, T. K.; Hejndorf, S.; Ng, Y. Z.; Low, J. Z. Y.; Rossant, J. Tryptophan-metabolizing gut microbes regulate adult neurogenesis via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. U. S. A. 2021, 118, e2021091118,  DOI: 10.1073/pnas.2021091118
    243. 243
      Kang, D.-W.; Adams, J. B.; Gregory, A. C.; Borody, T.; Chittick, L.; Fasano, A.; Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 2017, 5, 10,  DOI: 10.1186/s40168-016-0225-7
    244. 244
      Korpela, K.; Helve, O.; Kolho, K. L.; Saisto, T.; Skogberg, K.; Dikareva, E.; Stefanovic, V.; Salonen, A.; Andersson, S.; de Vos, W. M. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell 2020, 183, 324334.e325,  DOI: 10.1016/j.cell.2020.08.047
    245. 245
      Wikoff, W. R.; Anfora, A. T.; Liu, J.; Schultz, P. G.; Lesley, S. A.; Peters, E. C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 36983703,  DOI: 10.1073/pnas.0812874106
    246. 246
      Dodd, D.; Spitzer, M. H.; Van Treuren, W.; Merrill, B. D.; Hryckowian, A. J.; Higginbottom, S. K.; Le, A.; Cowan, T. M.; Nolan, G. P.; Fischbach, M. A. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648652,  DOI: 10.1038/nature24661
    247. 247
      Barrett, E.; Ross, R. P.; O’Toole, P. W.; Fitzgerald, G. F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411417,  DOI: 10.1111/j.1365-2672.2012.05344.x
    248. 248
      Asano, Y.; Hiramoto, T.; Nishino, R.; Aiba, Y.; Kimura, T.; Yoshihara, K.; Koga, Y.; Sudo, N. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G12881295,  DOI: 10.1152/ajpgi.00341.2012
    249. 249
      Pocivavsek, A.; Wu, H.-Q.; Potter, M. C.; Elmer, G. I.; Pellicciari, R.; Schwarcz, R. Fluctuations in Endogenous Kynurenic Acid Control Hippocampal Glutamate and Memory. Neuropsychopharmacology 2011, 36, 23572367,  DOI: 10.1038/npp.2011.127
    250. 250
      Potter, M. C.; Elmer, G. I.; Bergeron, R.; Albuquerque, E. X.; Guidetti, P.; Wu, H.-Q.; Schwarcz, R. Reduction of Endogenous Kynurenic Acid Formation Enhances Extracellular Glutamate, Hippocampal Plasticity, and Cognitive Behavior. Neuropsychopharmacology 2010, 35, 17341742,  DOI: 10.1038/npp.2010.39
    251. 251
      Reigstad, C. S.; Salmonson, C. E.; Rainey, J. F., III; Szurszewski, J. H.; Linden, D. R.; Sonnenburg, J. L.; Farrugia, G.; Kashyap, P. C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 13951403,  DOI: 10.1096/fj.14-259598
    252. 252
      Yano, J. M.; Yu, K.; Donaldson, G. P.; Shastri, G. G.; Ann, P.; Ma, L.; Nagler, C. R.; Ismagilov, R. F.; Mazmanian, S. K.; Hsiao, E. Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264276,  DOI: 10.1016/j.cell.2015.02.047
    253. 253
      Bellono, N. W.; Bayrer, J. R.; Leitch, D. B.; Castro, J.; Zhang, C.; O’Donnell, T. A.; Brierley, S. M.; Ingraham, H. A.; Julius, D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017, 170, 185198.e16,  DOI: 10.1016/j.cell.2017.05.034
    254. 254
      Ye, L.; Bae, M.; Cassilly, C. D.; Jabba, S. V.; Thorpe, D. W.; Martin, A. M.; Lu, H. Y.; Wang, J.; Thompson, J. D.; Lickwar, C. R. Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways. Cell Host Microbe 2021, 29, 179196.e79,  DOI: 10.1016/j.chom.2020.11.011
    255. 255
      O’Hagan, C.; Li, J. V.; Marchesi, J. R.; Plummer, S.; Garaiova, I.; Good, M. A. Long-term multi-species Lactobacillus and Bifidobacterium dietary supplement enhances memory and changes regional brain metabolites in middle-aged rats. Neurobiol Learn Mem 2017, 144, 3647,  DOI: 10.1016/j.nlm.2017.05.015
    256. 256
      Mao, J.-H.; Kim, Y.-M.; Zhou, Y.-X.; Hu, D.; Zhong, C.; Chang, H.; Brislawn, C. J.; Fansler, S.; Langley, S.; Wang, Y. Genetic and metabolic links between the murine microbiome and memory. Microbiome 2020, 8, 53,  DOI: 10.1186/s40168-020-00817-w
    257. 257
      El Hayek, L.; Khalifeh, M.; Zibara, V.; Abi Assaad, R.; Emmanuel, N.; Karnib, N.; El-Ghandour, R.; Nasrallah, P.; Bilen, M.; Ibrahim, P. Lactate Mediates the Effects of Exercise on Learning and Memory through SIRT1-Dependent Activation of Hippocampal Brain-Derived Neurotrophic Factor (BDNF). J. Neurosci. 2019, 39, 23692382,  DOI: 10.1523/JNEUROSCI.1661-18.2019
    258. 258
      Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nature Reviews Gastroenterology & Hepatology 2019, 16, 461478,  DOI: 10.1038/s41575-019-0157-3
    259. 259
      van de Wouw, M.; Boehme, M.; Lyte, J. M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain–gut axis alterations. Journal of Physiology 2018, 596, 49234944,  DOI: 10.1113/JP276431
    260. 260
      Bernasconi, R.; Jones, R. S.; Bittiger, H.; Olpe, H. R.; Heid, J.; Martin, P.; Klein, M.; Loo, P.; Braunwalder, A.; Schmutz, M. Dose pipecolic acid interact with the central GABA-ergic system?. J. Neural Transm. 1986, 67, 175189,  DOI: 10.1007/BF01243346
    261. 261
      Takahama, K.; Hashimoto, T.; Wang, M. W.; Akaike, N.; Hitoshi, T.; Okano, Y.; Kasé, Y.; Miyata, T. Pipecolic acid enhancement of GABA response in single neurons of rat brain. Neuropharmacology 1986, 25, 339342,  DOI: 10.1016/0028-3908(86)90263-7
    262. 262
      Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L. G.; Kundu, P. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158263ra158,  DOI: 10.1126/scitranslmed.3009759
    263. 263
      Li, H.; Sun, J.; Wang, F.; Ding, G.; Chen, W.; Fang, R.; Yao, Y.; Pang, M.; Lu, Z. Q.; Liu, J. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016, 1642, 7078,  DOI: 10.1016/j.brainres.2016.03.031
    264. 264
      Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J. K.; Carding, S. R.; Glen, R. C.; McArthur, S. Microbiome–host systems interactions: protective effects of propionate upon the blood–brain barrier. Microbiome 2018, 6, 55,  DOI: 10.1186/s40168-018-0439-y
    265. 265
      Quinn, M.; McMillin, M.; Galindo, C.; Frampton, G.; Pae, H. Y.; DeMorrow, S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig. Liver Dis. 2014, 46, 527534,  DOI: 10.1016/j.dld.2014.01.159
    266. 266
      Palmela, I.; Correia, L.; Silva, R. F. M.; Sasaki, H.; Kim, K. S.; Brites, D.; Brito, M. A. Hydrophilic bile acids protect human blood-brain barrier endothelial cells from disruption by unconjugated bilirubin: an in vitro study. Front. Neurosci. 2015, 9, 80,  DOI: 10.3389/fnins.2015.00080
    267. 267
      Hoyles, L.; Pontifex, M. G.; Rodriguez-Ramiro, I.; Anis-Alavi, M. A.; Jelane, K. S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A. L.; Carding, S. R. Regulation of blood–brain barrier integrity by microbiome-associated methylamines and cognition by trimethylamine N-oxide. Microbiome 2021, 9, 235,  DOI: 10.1186/s40168-021-01181-z
    268. 268
      Magistretti, P. J.; Allaman, I. A cellular perspective on brain energy metabolism and functional imaging. Neuron 2015, 86, 883901,  DOI: 10.1016/j.neuron.2015.03.035
    269. 269
      Wyss, M. T.; Jolivet, R.; Buck, A.; Magistretti, P. J.; Weber, B. In Vivo Evidence for Lactate as a Neuronal Energy Source. J. Neurosci. 2011, 31, 74777485,  DOI: 10.1523/JNEUROSCI.0415-11.2011
    270. 270
      Liu, Z.; Dai, X.; Zhang, H.; Shi, R.; Hui, Y.; Jin, X.; Zhang, W.; Wang, L.; Wang, Q.; Wang, D. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat. Commun. 2020, 11, 855,  DOI: 10.1038/s41467-020-14676-4
    271. 271
      Russell, W. R.; Hoyles, L.; Flint, H. J.; Dumas, M. E. Colonic bacterial metabolites and human health. Curr. Opin. Microbiol. 2013, 16, 246254,  DOI: 10.1016/j.mib.2013.07.002
    272. 272
      Ren, Z.; Zhang, R.; Li, Y.; Li, Y.; Yang, Z.; Yang, H. Ferulic acid exerts neuroprotective effects against cerebral ischemia/reperfusion-induced injury via antioxidant and anti-apoptotic mechanisms in vitro and in vivo. Int. J. Mol. Med. 2017, 40, 14441456,  DOI: 10.3892/ijmm.2017.3127
    273. 273
      Zeni, A. L. B.; Camargo, A.; Dalmagro, A. P. Ferulic acid reverses depression-like behavior and oxidative stress induced by chronic corticosterone treatment in mice. Steroids 2017, 125, 131136,  DOI: 10.1016/j.steroids.2017.07.006
    274. 274
      Verzelloni, E.; Pellacani, C.; Tagliazucchi, D.; Tagliaferri, S.; Calani, L.; Costa, L. G.; Brighenti, F.; Borges, G.; Crozier, A.; Conte, A. Antiglycative and neuroprotective activity of colon-derived polyphenol catabolites. Mol. Nutr. Food Res. 2011, 55, S35S43,  DOI: 10.1002/mnfr.201000525
    275. 275
      Macfarlane, S.; Macfarlane, G. T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 6772,  DOI: 10.1079/PNS2002207
    276. 276
      Thomas, C. M.; Hong, T.; van Pijkeren, J. P.; Hemarajata, P.; Trinh, D. V.; Hu, W.; Britton, R. A.; Kalkum, M.; Versalovic, J. Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PLoS One 2012, 7, e31951,  DOI: 10.1371/journal.pone.0031951
    277. 277
      De Biase, D.; Pennacchietti, E. Glutamate decarboxylase-dependent acid resistance in orally acquired bacteria: function, distribution and biomedical implications of the gadBC operon. Mol. Microbiol. 2012, 86, 770786,  DOI: 10.1111/mmi.12020
    278. 278
      Zhou, L.; Zeng, Y.; Zhang, H.; Ma, Y. The Role of Gastrointestinal Microbiota in Functional Dyspepsia: A Review. Front. Physiol. 2022, 13, 910568,  DOI: 10.3389/fphys.2022.910568
    279. 279
      Alexandrov, P. N.; Hill, J. M.; Zhao, Y.; Bond, T.; Taylor, C. M.; Percy, M. E.; Li, W.; Lukiw, W. J. Aluminum-induced generation of lipopolysaccharide (LPS) from the human gastrointestinal (GI)-tract microbiome-resident Bacteroides fragilis. J. Inorg. Biochem. 2020, 203, 110886,  DOI: 10.1016/j.jinorgbio.2019.110886
    280. 280
      Brown, E. M.; Ke, X.; Hitchcock, D.; Jeanfavre, S.; Avila-Pacheco, J.; Nakata, T.; Arthur, T. D.; Fornelos, N.; Heim, C.; Franzosa, E. A. Bacteroides-Derived Sphingolipids Are Critical for Maintaining Intestinal Homeostasis and Symbiosis. Cell Host Microbe 2019, 25, 668680.e67,  DOI: 10.1016/j.chom.2019.04.002
    281. 281
      Gao, K.; Pi, Y.; Mu, C. L.; Farzi, A.; Liu, Z.; Zhu, W. Y. Increasing carbohydrate availability in the hindgut promotes hypothalamic neurotransmitter synthesis: aromatic amino acids linking the microbiota-brain axis. J. Neurochem. 2019, 149, 641659,  DOI: 10.1111/jnc.14709
    282. 282
      Gao, K.; Mu, C.-l.; Farzi, A.; Zhu, W.-y. Tryptophan Metabolism: A Link Between the Gut Microbiota and Brain. Adv. Nutr. 2020, 11, 709723,  DOI: 10.1093/advances/nmz127
    283. 283
      Xiao, L.; Liu, Q.; Luo, M.; Xiong, L. Gut Microbiota-Derived Metabolites in Irritable Bowel Syndrome. Frontiers in Cellular and Infection Microbiology 2021, 11, 729346,  DOI: 10.3389/fcimb.2021.729346
    284. 284
      Mawe, G. M.; Hoffman, J. M. Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473486,  DOI: 10.1038/nrgastro.2013.105
    285. 285
      Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A. N.; Mackay, C. R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91119,  DOI: 10.1016/B978-0-12-800100-4.00003-9
    286. 286
      Felizardo, R. J. F.; Watanabe, I. K. M.; Dardi, P.; Rossoni, L. V.; Câmara, N. O. S. The interplay among gut microbiota, hypertension and kidney diseases: The role of short-chain fatty acids. Pharmacol. Res. 2019, 141, 366377,  DOI: 10.1016/j.phrs.2019.01.019
    287. 287
      Canfora, E. E.; Meex, R. C. R.; Venema, K.; Blaak, E. E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 2019, 15, 261273,  DOI: 10.1038/s41574-019-0156-z
    288. 288
      Tahara, Y.; Yamazaki, M.; Sukigara, H.; Motohashi, H.; Sasaki, H.; Miyakawa, H.; Haraguchi, A.; Ikeda, Y.; Fukuda, S.; Shibata, S. Gut Microbiota-Derived Short Chain Fatty Acids Induce Circadian Clock Entrainment in Mouse Peripheral Tissue. Sci. Rep. 2018, 8, 1395,  DOI: 10.1038/s41598-018-19836-7
    289. 289
      Feng, W.; Ao, H.; Peng, C. Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Front. Pharmacol. 2018, 9, 1354,  DOI: 10.3389/fphar.2018.01354
    290. 290
      Pan, L. L.; Li, B. B.; Pan, X. H.; Sun, J. Gut microbiota in pancreatic diseases: possible new therapeutic strategies. Acta Pharmacol. Sin. 2021, 42, 10271039,  DOI: 10.1038/s41401-020-00532-0
    291. 291
      Perino, A.; Demagny, H.; Velazquez-Villegas, L.; Schoonjans, K. Molecular Physiology of Bile Acid Signaling in Health, Disease, and Aging. Physiol. Rev. 2021, 101, 683731,  DOI: 10.1152/physrev.00049.2019
    292. 292
      Wahlström, A.; Sayin, S. I.; Marschall, H. U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 4150,  DOI: 10.1016/j.cmet.2016.05.005
    293. 293
      Poland, J. C.; Flynn, C. R. Bile Acids, Their Receptors, and the Gut Microbiota. Physiology (Bethesda) 2021, 36, 235245,  DOI: 10.1152/physiol.00028.2020
    294. 294
      Govindarajan, K.; MacSharry, J.; Casey, P. G.; Shanahan, F.; Joyce, S. A.; Gahan, C. G. M. Unconjugated Bile Acids Influence Expression of Circadian Genes: A Potential Mechanism for Microbe-Host Crosstalk. PLoS One 2016, 11, e0167319  DOI: 10.1371/journal.pone.0167319
    295. 295
      McMillin, M.; DeMorrow, S. Effects of bile acids on neurological function and disease. FASEB J. 2016, 30, 36583668,  DOI: 10.1096/fj.201600275R
    296. 296
      Roth, W.; Zadeh, K.; Vekariya, R.; Ge, Y.; Mohamadzadeh, M. Tryptophan Metabolism and Gut-Brain Homeostasis. Int. J. Mol. Sci. 2021, 22, 2973,  DOI: 10.3390/ijms22062973
    297. 297
      Zhang, J.; Zhu, S.; Ma, N.; Johnston, L. J.; Wu, C.; Ma, X. Metabolites of microbiota response to tryptophan and intestinal mucosal immunity: A therapeutic target to control intestinal inflammation. Med. Res. Rev. 2021, 41, 10611088,  DOI: 10.1002/med.21752
    298. 298
      Modoux, M.; Rolhion, N.; Mani, S.; Sokol, H. Tryptophan Metabolism as a Pharmacological Target. Trends Pharmacol. Sci. 2021, 42, 6073,  DOI: 10.1016/j.tips.2020.11.006
    299. 299
      Roager, H. M.; Licht, T. R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294,  DOI: 10.1038/s41467-018-05470-4
    300. 300
      Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716724,  DOI: 10.1016/j.chom.2018.05.003
    301. 301
      Wang, Z.; Bergeron, N.; Levison, B. S.; Li, X. S.; Chiu, S.; Jia, X.; Koeth, R. A.; Li, L.; Wu, Y.; Tang, W. H. W. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583594,  DOI: 10.1093/eurheartj/ehy799
    302. 302
      Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: how gut microbiota contributes to heart failure. Transl. Res. 2021, 228, 109125,  DOI: 10.1016/j.trsl.2020.08.007
    303. 303
      Yang, S.; Li, X.; Yang, F.; Zhao, R.; Pan, X.; Liang, J.; Tian, L.; Li, X.; Liu, L.; Xing, Y. Gut Microbiota-Dependent Marker TMAO in Promoting Cardiovascular Disease: Inflammation Mechanism, Clinical Prognostic, and Potential as a Therapeutic Target. Front. Pharmacol. 2019, 10, 1360,  DOI: 10.3389/fphar.2019.01360
    304. 304
      Rudzki, L.; Stone, T. W.; Maes, M.; Misiak, B.; Samochowiec, J.; Szulc, A. Gut microbiota-derived vitamins - underrated powers of a multipotent ally in psychiatric health and disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 107, 110240,  DOI: 10.1016/j.pnpbp.2020.110240
    305. 305
      Stacchiotti, V.; Rezzi, S.; Eggersdorfer, M.; Galli, F. Metabolic and functional interplay between gut microbiota and fat-soluble vitamins. Crit Rev. Food Sci. Nutr 2021, 61, 32113232,  DOI: 10.1080/10408398.2020.1793728
    306. 306
      Masri, O. A.; Chalhoub, J. M.; Sharara, A. I. Role of vitamins in gastrointestinal diseases. World J. Gastroenterol. 2015, 21, 51915209,  DOI: 10.3748/wjg.v21.i17.5191
    307. 307
      McCarville, J. L.; Chen, G. Y.; Cuevas, V. D.; Troha, K.; Ayres, J. S. Microbiota Metabolites in Health and Disease. Annu. Rev. Immunol. 2020, 38, 147170,  DOI: 10.1146/annurev-immunol-071219-125715
    308. 308
      Wang, Y.; Li, N.; Yang, J. J.; Zhao, D. M.; Chen, B.; Zhang, G. Q.; Chen, S.; Cao, R. F.; Yu, H.; Zhao, C. Y. Probiotics and fructo-oligosaccharide intervention modulate the microbiota-gut brain axis to improve autism spectrum reducing also the hyper-serotonergic state and the dopamine metabolism disorder. Pharmacol. Res. 2020, 157, 104784,  DOI: 10.1016/j.phrs.2020.104784
    309. 309
      Morais, L. H.; Schreiber, H. L. t.; Mazmanian, S. K. The gut microbiota-brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241255,  DOI: 10.1038/s41579-020-00460-0
    310. 310
      Cani, P. D.; Amar, J.; Iglesias, M. A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A. M.; Fava, F.; Tuohy, K. M.; Chabo, C. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 17611772,  DOI: 10.2337/db06-1491
    311. 311
      Serino, M.; Luche, E.; Gres, S.; Baylac, A.; Bergé, M.; Cenac, C.; Waget, A.; Klopp, P.; Iacovoni, J.; Klopp, C. Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 2012, 61, 543553,  DOI: 10.1136/gutjnl-2011-301012
    312. 312
      Ostojic, S. M. Inadequate Production of H(2) by Gut Microbiota and Parkinson Disease. Trends Endocrinol Metab 2018, 29, 286288,  DOI: 10.1016/j.tem.2018.02.006
    313. 313
      Kalantar-Zadeh, K.; Berean, K. J.; Burgell, R. E.; Muir, J. G.; Gibson, P. R. Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 733747,  DOI: 10.1038/s41575-019-0193-z
    314. 314
      Pacher, P.; Beckman, J. S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315424,  DOI: 10.1152/physrev.00029.2006
    315. 315
      Singh, S. B.; Lin, H. C. Hydrogen Sulfide in Physiology and Diseases of the Digestive Tract. Microorganisms 2015, 3, 866889,  DOI: 10.3390/microorganisms3040866
    316. 316
      Sen, N. Functional and Molecular Insights of Hydrogen Sulfide Signaling and Protein Sulfhydration. J. Mol. Biol. 2017, 429, 543561,  DOI: 10.1016/j.jmb.2016.12.015
    317. 317
      Skonieczna-Żydecka, K.; Marlicz, W.; Misera, A.; Koulaouzidis, A.; Łoniewski, I. Microbiome─The Missing Link in the Gut-Brain Axis: Focus on Its Role in Gastrointestinal and Mental Health. Journal of Clinical Medicine 2018, 7, 521,  DOI: 10.3390/jcm7120521
    318. 318
      Schmulson, M. J.; Drossman, D. A. What Is New in Rome IV. J. Neurogastroenterol. Motil. 2017, 23, 151163,  DOI: 10.5056/jnm16214
    319. 319
      Stasi, C.; Nisita, C.; Cortopassi, S.; Corretti, G.; Gambaccini, D.; De Bortoli, N.; Fani, B.; Simonetti, N.; Ricchiuti, A.; Dell’Osso, L. Subthreshold Psychiatric Psychopathology in Functional Gastrointestinal Disorders: Can It Be the Bridge between Gastroenterology and Psychiatry?. Gastroenterol. Res. Pract. 2017, 2017, 1953435,  DOI: 10.1155/2017/1953435
    320. 320
      Cryan, J. F.; O’Riordan, K. J.; Cowan, C. S. M.; Sandhu, K. V.; Bastiaanssen, T. F. S.; Boehme, M.; Codagnone, M. G.; Cussotto, S.; Fulling, C.; Golubeva, A. V. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 18772013,  DOI: 10.1152/physrev.00018.2018
    321. 321
      Gralnek, I. M.; Hays, R. D.; Kilbourne, A.; Naliboff, B.; Mayer, E. A. The impact of irritable bowel syndrome on health-related quality of life. Gastroenterology 2000, 119, 654660,  DOI: 10.1053/gast.2000.16484
    322. 322
      O’Keeffe, M.; Lomer, M. C. Who should deliver the low FODMAP diet and what educational methods are optimal: a review. J. Gastroenterol. Hepatol. 2017, 32, 2326,  DOI: 10.1111/jgh.13690
    323. 323
      Koloski, N. A.; Jones, M.; Talley, N. J. Evidence that independent gut-to-brain and brain-to-gut pathways operate in the irritable bowel syndrome and functional dyspepsia: a 1-year population-based prospective study. Alimentary pharmacology & therapeutics 2016, 44, 592600,  DOI: 10.1111/apt.13738
    324. 324
      Eijsbouts, C.; Zheng, T.; Kennedy, N. A.; Bonfiglio, F.; Anderson, C. A.; Moutsianas, L.; Holliday, J.; Shi, J.; Shringarpure, S.; Agee, M. Genome-wide analysis of 53,400 people with irritable bowel syndrome highlights shared genetic pathways with mood and anxiety disorders. Nat. Genet. 2021, 53, 15431552,  DOI: 10.1038/s41588-021-00950-8
    325. 325
      Schwarz, E.; Maukonen, J.; Hyytiäinen, T.; Kieseppä, T.; Orešič, M.; Sabunciyan, S.; Mantere, O.; Saarela, M.; Yolken, R.; Suvisaari, J. Analysis of microbiota in first episode psychosis identifies preliminary associations with symptom severity and treatment response. Schizophr. Res. 2018, 192, 398403,  DOI: 10.1016/j.schres.2017.04.017
    326. 326
      Castro-Nallar, E.; Bendall, M. L.; Pérez-Losada, M.; Sabuncyan, S.; Severance, E. G.; Dickerson, F. B.; Schroeder, J. R.; Yolken, R. H.; Crandall, K. A. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ. 2015, 3, e1140,  DOI: 10.7717/peerj.1140
    327. 327
      Belkaid, Y.; Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121141,  DOI: 10.1016/j.cell.2014.03.011
    328. 328
      Anderson, G.; Maes, M. Bipolar disorder: role of immune-inflammatory cytokines, oxidative and nitrosative stress and tryptophan catabolites. Curr. Psychiatry Rep 2015, 17, 8,  DOI: 10.1007/s11920-014-0541-1
    329. 329
      Clarke, G.; Fitzgerald, P.; Cryan, J. F.; Cassidy, E. M.; Quigley, E. M.; Dinan, T. G. Tryptophan degradation in irritable bowel syndrome: evidence of indoleamine 2,3-dioxygenase activation in a male cohort. BMC Gastroenterol. 2009, 9, 6,  DOI: 10.1186/1471-230X-9-6
    330. 330
      Clarke, G.; McKernan, D.; Gaszner, G.; Quigley, E.; Cryan, J.; Dinan, T. A Distinct Profile of Tryptophan Metabolism along the Kynurenine Pathway Downstream of Toll-Like Receptor Activation in Irritable Bowel Syndrome. Front. Pharmacol. 2012, 3, 90,  DOI: 10.3389/fphar.2012.00090
    331. 331
      Liang, X.; FitzGerald, G. A. Timing the Microbes: The Circadian Rhythm of the Gut Microbiome. J. Biol. Rhythms 2017, 32, 505515,  DOI: 10.1177/0748730417729066
    332. 332
      Enck, P.; Aziz, Q.; Barbara, G.; Farmer, A. D.; Fukudo, S.; Mayer, E. A.; Niesler, B.; Quigley, E. M.; Rajilić-Stojanović, M.; Schemann, M. Irritable bowel syndrome. Nat. Rev. Dis Primers 2016, 2, 16014,  DOI: 10.1038/nrdp.2016.14
    333. 333
      Dalile, B.; Vervliet, B.; Bergonzelli, G.; Verbeke, K.; Van Oudenhove, L. Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: a randomized, placebo-controlled trial. Neuropsychopharmacology 2020, 45, 22572266,  DOI: 10.1038/s41386-020-0732-x
    334. 334
      Bercik, P.; Park, A. J.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; Deng, Y.; Blennerhassett, P. A.; Fahnestock, M.; Moine, D. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol. Motil. 2011, 23, 11321139,  DOI: 10.1111/j.1365-2982.2011.01796.x
    335. 335
      Mishima, Y.; Ishihara, S. Molecular Mechanisms of Microbiota-Mediated Pathology in Irritable Bowel Syndrome. Int. J. Mol. Sci. 2020, 21, 8664,  DOI: 10.3390/ijms21228664
    336. 336
      Leeming, E. R.; Johnson, A. J.; Spector, T. D.; Le Roy, C. I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862,  DOI: 10.3390/nu11122862
    337. 337
      Hillestad, E. M. R.; van der Meeren, A.; Nagaraja, B. H.; Bjo̷rsvik, B. R.; Haleem, N.; Benitez-Paez, A.; Sanz, Y.; Hausken, T.; Lied, G. A.; Lundervold, A. Gut bless you: The microbiota-gut-brain axis in irritable bowel syndrome. World J. Gastroenterol. 2022, 28, 412431,  DOI: 10.3748/wjg.v28.i4.412
    338. 338
      Hugerth, L. W.; Andreasson, A.; Talley, N. J.; Forsberg, A. M.; Kjellström, L.; Schmidt, P. T.; Agreus, L.; Engstrand, L. No distinct microbiome signature of irritable bowel syndrome found in a Swedish random population. Gut 2020, 69, 10761084,  DOI: 10.1136/gutjnl-2019-318717
    339. 339
      Sundin, J.; Aziz, I.; Nordlander, S.; Polster, A.; Hu, Y. O. O.; Hugerth, L. W.; Pennhag, A. A. L.; Engstrand, L.; Törnblom, H.; Simrén, M. Evidence of altered mucosa-associated and fecal microbiota composition in patients with Irritable Bowel Syndrome. Sci. Rep. 2020, 10, 593,  DOI: 10.1038/s41598-020-57468-y
    340. 340
      Mayer, E. A.; Labus, J.; Aziz, Q.; Tracey, I.; Kilpatrick, L.; Elsenbruch, S.; Schweinhardt, P.; Van Oudenhove, L.; Borsook, D. Role of brain imaging in disorders of brain-gut interaction: a Rome Working Team Report. Gut 2019, 68, 17011715,  DOI: 10.1136/gutjnl-2019-318308
    341. 341
      Seminowicz, D. A.; Labus, J. S.; Bueller, J. A.; Tillisch, K.; Naliboff, B. D.; Bushnell, M. C.; Mayer, E. A. Regional gray matter density changes in brains of patients with irritable bowel syndrome. Gastroenterology 2010, 139, 4857.e42,  DOI: 10.1053/j.gastro.2010.03.049
    342. 342
      Kano, M.; Grinsvall, C.; Ran, Q.; Dupont, P.; Morishita, J.; Muratsubaki, T.; Mugikura, S.; Ly, H. G.; Törnblom, H.; Ljungberg, M. Resting state functional connectivity of the pain matrix and default mode network in irritable bowel syndrome: a graph theoretical analysis. Sci. Rep. 2020, 10, 11015,  DOI: 10.1038/s41598-020-67048-9
    343. 343
      Wilder-Smith, C. H.; Schindler, D.; Lovblad, K.; Redmond, S. M.; Nirkko, A. Brain functional magnetic resonance imaging of rectal pain and activation of endogenous inhibitory mechanisms in irritable bowel syndrome patient subgroups and healthy controls. Gut 2004, 53, 15951601,  DOI: 10.1136/gut.2003.028514
    344. 344
      Tillisch, K.; Mayer, E. A.; Labus, J. S. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology 2011, 140, 91100,  DOI: 10.1053/j.gastro.2010.07.053
    345. 345
      Ringel, Y.; Drossman, D. A.; Leserman, J. L.; Suyenobu, B. Y.; Wilber, K.; Lin, W.; Whitehead, W. E.; Naliboff, B. D.; Berman, S.; Mayer, E. A. Effect of abuse history on pain reports and brain responses to aversive visceral stimulation: an FMRI study. Gastroenterology 2008, 134, 396404,  DOI: 10.1053/j.gastro.2007.11.011
    346. 346
      Mayer, E. A.; Bradesi, S.; Chang, L.; Spiegel, B. M.; Bueller, J. A.; Naliboff, B. D. Functional GI disorders: from animal models to drug development. Gut 2008, 57, 384404,  DOI: 10.1136/gut.2006.101675
    347. 347
      Labus, J. S.; Osadchiy, V.; Hsiao, E. Y.; Tap, J.; Derrien, M.; Gupta, A.; Tillisch, K.; Le Nevé, B.; Grinsvall, C.; Ljungberg, M. Evidence for an association of gut microbial Clostridia with brain functional connectivity and gastrointestinal sensorimotor function in patients with irritable bowel syndrome, based on tripartite network analysis. Microbiome 2019, 7, 45,  DOI: 10.1186/s40168-019-0656-z
    348. 348
      Osadchiy, V.; Mayer, E. A.; Gao, K.; Labus, J. S.; Naliboff, B.; Tillisch, K.; Chang, L.; Jacobs, J. P.; Hsiao, E. Y.; Gupta, A. Analysis of brain networks and fecal metabolites reveals brain-gut alterations in premenopausal females with irritable bowel syndrome. Transl Psychiatry 2020, 10, 367,  DOI: 10.1038/s41398-020-01071-2
    349. 349
      Sarnoff, R. P.; Bhatt, R. R.; Osadchiy, V.; Dong, T.; Labus, J. S.; Kilpatrick, L. A.; Chen, Z.; Subramanyam, V.; Zhang, Y.; Ellingson, B. M. A multi-omic brain gut microbiome signature differs between IBS subjects with different bowel habits. Neuropharmacology 2023, 225, 109381,  DOI: 10.1016/j.neuropharm.2022.109381
    350. 350
      Berman, S. M.; Naliboff, B. D.; Suyenobu, B.; Labus, J. S.; Stains, J.; Ohning, G.; Kilpatrick, L.; Bueller, J. A.; Ruby, K.; Jarcho, J. Reduced brainstem inhibition during anticipated pelvic visceral pain correlates with enhanced brain response to the visceral stimulus in women with irritable bowel syndrome. J. Neurosci. 2008, 28, 349359,  DOI: 10.1523/JNEUROSCI.2500-07.2008
    351. 351
      Guleria, A.; Karyampudi, A.; Singh, R.; Khetrapal, C. L.; Verma, A.; Ghoshal, U. C.; Kumar, D. Mapping of Brain Activations to Rectal Balloon Distension Stimuli in Male Patients with Irritable Bowel Syndrome Using Functional Magnetic Resonance Imaging. J. Neurogastroenterol. Motil. 2017, 23, 415427,  DOI: 10.5056/jnm16148
    352. 352
      Ticinesi, A.; Milani, C.; Lauretani, F.; Nouvenne, A.; Mancabelli, L.; Lugli, G. A.; Turroni, F.; Duranti, S.; Mangifesta, M.; Viappiani, A. Gut microbiota composition is associated with polypharmacy in elderly hospitalized patients. Sci. Rep. 2017, 7, 11102,  DOI: 10.1038/s41598-017-10734-y
    353. 353
      Chopra, K.; Kumar, B.; Kuhad, A. Pathobiological targets of depression. Expert Opin. Ther. Targets 2011, 15, 379400,  DOI: 10.1517/14728222.2011.553603
    354. 354
      Dinan, T. G.; Cryan, J. F. Gut-brain axis in 2016: Brain-gut-microbiota axis - mood, metabolism and behaviour. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 6970,  DOI: 10.1038/nrgastro.2016.200
    355. 355
      Foster, J. A.; McVey Neufeld, K. A. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305312,  DOI: 10.1016/j.tins.2013.01.005
    356. 356
      Halkjær, S. I.; Christensen, A. H.; Lo, B. Z. S.; Browne, P. D.; Günther, S.; Hansen, L. H.; Petersen, A. M. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut 2018, 67, 21072115,  DOI: 10.1136/gutjnl-2018-316434
    357. 357
      Holvoet, T.; Joossens, M.; Wang, J.; Boelens, J.; Verhasselt, B.; Laukens, D.; van Vlierberghe, H.; Hindryckx, P.; De Vos, M.; De Looze, D. Assessment of faecal microbial transfer in irritable bowel syndrome with severe bloating. Gut 2017, 66, 980982,  DOI: 10.1136/gutjnl-2016-312513
    358. 358
      Mizuno, S.; Masaoka, T.; Naganuma, M.; Kishimoto, T.; Kitazawa, M.; Kurokawa, S.; Nakashima, M.; Takeshita, K.; Suda, W.; Mimura, M. Bifidobacterium-Rich Fecal Donor May Be a Positive Predictor for Successful Fecal Microbiota Transplantation in Patients with Irritable Bowel Syndrome. Digestion 2017, 96, 2938,  DOI: 10.1159/000471919
    359. 359
      Zoller, V.; Laguna, A. L.; Prazeres Da Costa, O.; Buch, T.; Göke, B.; Storr, M. Fecal microbiota transfer (FMT) in a patient with refractory irritable bowel syndrome. Dtsch. Med. Wochenschr. 2015, 140, 12321236,  DOI: 10.1055/s-0041-103798
    360. 360
      Drago, L.; Meroni, G.; Pistone, D.; Pasquale, L.; Milazzo, G.; Monica, F.; Aragona, S.; Ficano, L.; Vassallo, R. Evaluation of main functional dyspepsia symptoms after probiotic administration in patients receiving conventional pharmacological therapies. J. Int. Med. Res. 2021, 49, 300060520982657,  DOI: 10.1177/0300060520982657
    361. 361
      Potter, M. D. E.; Talley, N. J. Editorial: new insights into the global prevalence of uninvestigated and functional dyspepsia. Aliment. Pharmacol. Ther. 2020, 52 (8), 14071408,  DOI: 10.1111/apt.16059
    362. 362
      Zhong, L.; Shanahan, E. R.; Raj, A.; Koloski, N. A.; Fletcher, L.; Morrison, M.; Walker, M. M.; Talley, N. J.; Holtmann, G. Dyspepsia and the microbiome: time to focus on the small intestine. Gut 2017, 66, 11681169,  DOI: 10.1136/gutjnl-2016-312574
    363. 363
      Vanheel, H.; Vicario, M.; Vanuytsel, T.; Van Oudenhove, L.; Martinez, C.; Keita, Å. V.; Pardon, N.; Santos, J.; Söderholm, J. D.; Tack, J. Impaired duodenal mucosal integrity and low-grade inflammation in functional dyspepsia. Gut 2014, 63, 262271,  DOI: 10.1136/gutjnl-2012-303857
    364. 364
      Liu, X. J.; Xie, W. R.; Wu, L. H.; Ye, Z. N.; Zhang, X. Y.; Zhang, R.; He, X. X. Changes in oral flora of patients with functional dyspepsia. Sci. Rep. 2021, 11, 8089,  DOI: 10.1038/s41598-021-87600-5
    365. 365
      Aro, P.; Talley, N. J.; Johansson, S. E.; Agréus, L.; Ronkainen, J. Anxiety Is Linked to New-Onset Dyspepsia in the Swedish Population: A 10-Year Follow-up Study. Gastroenterology 2015, 148, 928937,  DOI: 10.1053/j.gastro.2015.01.039
    366. 366
      Van Oudenhove, L.; Aziz, Q. The role of psychosocial factors and psychiatric disorders in functional dyspepsia. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 158167,  DOI: 10.1038/nrgastro.2013.10
    367. 367
      Hannibal, K. E.; Bishop, M. D. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys. Ther. 2014, 94, 18161825,  DOI: 10.2522/ptj.20130597
    368. 368
      Koloski, N. A.; Jones, M.; Kalantar, J.; Weltman, M.; Zaguirre, J.; Talley, N. J. The brain--gut pathway in functional gastrointestinal disorders is bidirectional: a 12-year prospective population-based study. Gut 2012, 61, 12841290,  DOI: 10.1136/gutjnl-2011-300474
    369. 369
      Cryan, J. F.; Dinan, T. G. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701712,  DOI: 10.1038/nrn3346
    370. 370
      Browning, K. N.; Travagli, R. A. Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. Compr Physiol 2014, 4, 13391368,  DOI: 10.1002/cphy.c130055
    371. 371
      Barberio, B.; Judge, C.; Savarino, E. V.; Ford, A. C. Global prevalence of functional constipation according to the Rome criteria: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2021, 6, 638648,  DOI: 10.1016/S2468-1253(21)00111-4
    372. 372
      Vriesman, M. H.; Koppen, I. J. N.; Camilleri, M.; Di Lorenzo, C.; Benninga, M. A. Management of functional constipation in children and adults. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 2139,  DOI: 10.1038/s41575-019-0222-y
    373. 373
      Zhuang, M.; Shang, W.; Ma, Q.; Strappe, P.; Zhou, Z. Abundance of Probiotics and Butyrate-Production Microbiome Manages Constipation via Short-Chain Fatty Acids Production and Hormones Secretion. Mol. Nutr. Food Res. 2019, 63, e1801187,  DOI: 10.1002/mnfr.201801187
    374. 374
      Attaluri, A.; Jackson, M.; Valestin, J.; Rao, S. S. Methanogenic flora is associated with altered colonic transit but not stool characteristics in constipation without IBS. Am. J. Gastroenterol. 2010, 105, 14071411,  DOI: 10.1038/ajg.2009.655
    375. 375
      Chen, Y.; Wu, T.; Lu, W.; Yuan, W.; Pan, M.; Lee, Y. K.; Zhao, J.; Zhang, H.; Chen, W.; Zhu, J. Predicting the Role of the Human Gut Microbiome in Constipation Using Machine-Learning Methods: A Meta-Analysis. Microorganisms 2021, 9, 2149,  DOI: 10.3390/microorganisms9102149
    376. 376
      Tian, H.; Ye, C.; Yang, B.; Cui, J.; Zheng, Z.; Wu, C.; Zhou, S.; Lv, X.; Qin, N.; Qin, H. Gut Metagenome as a Potential Diagnostic and Predictive Biomarker in Slow Transit Constipation. Front. Med. 2022, 8, 777961,  DOI: 10.3389/fmed.2021.777961
    377. 377
      Sartor, R. B. Optimal sampling of the intestinal microbiota for research. Nature Reviews Gastroenterology & Hepatology 2015, 12, 253254,  DOI: 10.1038/nrgastro.2015.46
    378. 378
      Swidsinski, A.; Loening-Baucke, V.; Verstraelen, H.; Osowska, S.; Doerffel, Y. Biostructure of fecal microbiota in healthy subjects and patients with chronic idiopathic diarrhea. Gastroenterology 2008, 135, 568579,  DOI: 10.1053/j.gastro.2008.04.017
    379. 379
      Durbán, A.; Abellán, J. J.; Jiménez-Hernández, N.; Salgado, P.; Ponce, M.; Ponce, J.; Garrigues, V.; Latorre, A.; Moya, A. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome. Environ. Microbiol. Rep. 2012, 4, 242247,  DOI: 10.1111/j.1758-2229.2012.00327.x
    380. 380
      Parthasarathy, G.; Chen, J.; Chen, X.; Chia, N.; O’Connor, H. M.; Wolf, P. G.; Gaskins, H. R.; Bharucha, A. E. Relationship Between Microbiota of the Colonic Mucosa vs Feces and Symptoms, Colonic Transit, and Methane Production in Female Patients With Chronic Constipation. Gastroenterology 2016, 150, 367379.e1,  DOI: 10.1053/j.gastro.2015.10.005
    381. 381
      Procházková, N.; Falony, G.; Dragsted, L. O.; Licht, T. R.; Raes, J.; Roager, H. M. Advancing human gut microbiota research by considering gut transit time. Gut 2023, 72, 180191,  DOI: 10.1136/gutjnl-2022-328166
    382. 382
      Hosseinzadeh, S. T.; Poorsaadati, S.; Radkani, B.; Forootan, M. Psychological disorders in patients with chronic constipation. Gastroenterol. Hepatol. Bed Bench 2011, 4, 159163
    383. 383
      Duan, S.; Liu, L.; Li, G.; Wang, J.; Hu, Y.; Zhang, W.; Tan, Z.; Jia, Z.; Zhang, L.; von Deneen, K. M. Altered Functional Connectivity Within and Between Salience and Sensorimotor Networks in Patients With Functional Constipation. Front. Neurosci. 2021, 15, 628880,  DOI: 10.3389/fnins.2021.628880
    384. 384
      Hu, C.; Liu, L.; Liu, L.; Zhang, J.; Hu, Y.; Zhang, W.; Ding, Y.; Wang, Y.; Zhang, Z.; von Deneen, K. M. Cortical morphometry alterations in brain regions involved in emotional, motor-control and self-referential processing in patients with functional constipation. Brain Imaging Behav. 2020, 14, 18991907,  DOI: 10.1007/s11682-019-00133-4
    385. 385
      Jin, Q.; Duan, S.; Li, G.; Sun, L.; Hu, Y.; Hu, C.; Zhao, J.; von Deneen, K. M.; Qian, L.; Wang, H. Sex-related differences in resting-state brain activity and connectivity in the orbital frontal cortex and insula in patients with functional constipation. Neurogastroenterol. Motil. 2019, 31, e13566,  DOI: 10.1111/nmo.13566
    386. 386
      Li, G.; Zhang, W.; Hu, Y.; Wang, J.; Li, J.; Jia, Z.; Zhang, L.; Sun, L.; von Deneen, K. M.; Duan, S. Distinct Basal Brain Functional Activity and Connectivity in the Emotional-Arousal Network and Thalamus in Patients With Functional Constipation Associated With Anxiety and/or Depressive Disorders. Psychosom. Med. 2021, 83, 707714,  DOI: 10.1097/PSY.0000000000000958
    387. 387
      Liu, L.; Hu, C.; Hu, Y.; Zhang, W.; Zhang, Z.; Ding, Y.; Wang, Y.; von Deneen, K. M.; Sun, L.; Wang, H. Abnormalities in the thalamo-cortical network in patients with functional constipation. Brain Imaging Behav. 2021, 15, 630642,  DOI: 10.1007/s11682-020-00273-y
    388. 388
      Peihong, M.; Tao, Y.; Zhaoxuan, H.; Sha, Y.; Li, C.; Kunnan, X.; Jingwen, C.; Likai, H.; Yuke, T.; Yuyi, G. Alterations of White Matter Network Properties in Patients With Functional Constipation. Front. Neurol. 2021, 12, 627130,  DOI: 10.3389/fneur.2021.627130
    389. 389
      Zhu, Q.; Cai, W.; Zheng, J.; Li, G.; Meng, Q.; Liu, Q.; Zhao, J.; von Deneen, K. M.; Wang, Y.; Cui, G. Distinct resting-state brain activity in patients with functional constipation. Neurosci. Lett. 2016, 632, 141146,  DOI: 10.1016/j.neulet.2016.08.042
    390. 390
      Mayer, E. A. Gut feelings: the emerging biology of gut–brain communication. Nat. Rev. Neurosci. 2011, 12, 453466,  DOI: 10.1038/nrn3071
    391. 391
      Rubio, A.; Van Oudenhove, L.; Pellissier, S.; Ly, H. G.; Dupont, P.; Lafaye de Micheaux, H.; Tack, J.; Dantzer, C.; Delon-Martin, C.; Bonaz, B. Uncertainty in anticipation of uncomfortable rectal distension is modulated by the autonomic nervous system--a fMRI study in healthy volunteers. NeuroImage 2015, 107, 1022,  DOI: 10.1016/j.neuroimage.2014.11.043
    392. 392
      Molina-Torres, G.; Rodriguez-Arrastia, M.; Roman, P.; Sanchez-Labraca, N.; Cardona, D. Stress and the gut microbiota-brain axis. Behav. Pharmacol. 2019, 30, 187200,  DOI: 10.1097/FBP.0000000000000478
    393. 393
      Vanuytsel, T.; van Wanrooy, S.; Vanheel, H.; Vanormelingen, C.; Verschueren, S.; Houben, E.; Salim Rasoel, S.; Tóth, J.; Holvoet, L.; Farré, R. Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut 2014, 63, 12931299,  DOI: 10.1136/gutjnl-2013-305690
    394. 394
      Yarandi, S. S.; Peterson, D. A.; Treisman, G. J.; Moran, T. H.; Pasricha, P. J. Modulatory Effects of Gut Microbiota on the Central Nervous System: How Gut Could Play a Role in Neuropsychiatric Health and Diseases. J. Neurogastroenterol. Motil. 2016, 22, 201212,  DOI: 10.5056/jnm15146
    395. 395
      Kelly, J. R.; Kennedy, P. J.; Cryan, J. F.; Dinan, T. G.; Clarke, G.; Hyland, N. P. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392,  DOI: 10.3389/fncel.2015.00392
    396. 396
      Tannock, G. W.; Savage, D. C. Influences of dietary and environmental stress on microbial populations in the murine gastrointestinal tract. Infect. Immun. 1974, 9, 591598,  DOI: 10.1128/iai.9.3.591-598.1974
    397. 397
      Yang, H.; Zhao, X.; Tang, S.; Huang, H.; Zhao, X.; Ning, Z.; Fu, X.; Zhang, C. Probiotics reduce psychological stress in patients before laryngeal cancer surgery. Asia Pac. J. Clin. Oncol. 2016, 12, e9296,  DOI: 10.1111/ajco.12120
    398. 398
      Karl, J. P.; Margolis, L. M.; Madslien, E. H.; Murphy, N. E.; Castellani, J. W.; Gundersen, Y.; Hoke, A. V.; Levangie, M. W.; Kumar, R.; Chakraborty, N. Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G559g571,  DOI: 10.1152/ajpgi.00066.2017
    399. 399
      Kato-Kataoka, A.; Nishida, K.; Takada, M.; Kawai, M.; Kikuchi-Hayakawa, H.; Suda, K.; Ishikawa, H.; Gondo, Y.; Shimizu, K.; Matsuki, T. Fermented Milk Containing Lactobacillus casei Strain Shirota Preserves the Diversity of the Gut Microbiota and Relieves Abdominal Dysfunction in Healthy Medical Students Exposed to Academic Stress. Appl. Environ. Microbiol. 2016, 82, 36493658,  DOI: 10.1128/AEM.04134-15
    400. 400
      Takada, M.; Nishida, K.; Kataoka-Kato, A.; Gondo, Y.; Ishikawa, H.; Suda, K.; Kawai, M.; Hoshi, R.; Watanabe, O.; Igarashi, T. Probiotic Lactobacillus casei strain Shirota relieves stress-associated symptoms by modulating the gut-brain interaction in human and animal models. Neurogastroenterol. Motil. 2016, 28, 10271036,  DOI: 10.1111/nmo.12804
    401. 401
      Zijlmans, M. A.; Korpela, K.; Riksen-Walraven, J. M.; de Vos, W. M.; de Weerth, C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 2015, 53, 233245,  DOI: 10.1016/j.psyneuen.2015.01.006
    402. 402
      Naseribafrouei, A.; Hestad, K.; Avershina, E.; Sekelja, M.; Linlo̷kken, A.; Wilson, R.; Rudi, K. Correlation between the human fecal microbiota and depression. Neurogastroenterol. Motil. 2014, 26, 11551162,  DOI: 10.1111/nmo.12378
    403. 403
      Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J. Altered fecal microbiota composition in patients with major depressive disorder. Brain. Behav. Immun. 2015, 48, 186194,  DOI: 10.1016/j.bbi.2015.03.016
    404. 404
      Tillisch, K.; Mayer, E. A.; Gupta, A.; Gill, Z.; Brazeilles, R.; Le Nevé, B.; van Hylckama Vlieg, J. E. T.; Guyonnet, D.; Derrien, M.; Labus, J. S. Brain Structure and Response to Emotional Stimuli as Related to Gut Microbial Profiles in Healthy Women. Psychosom. Med. 2017, 79, 905913,  DOI: 10.1097/PSY.0000000000000493
    405. 405
      Pfau, M. L.; Russo, S. J. Peripheral and Central Mechanisms of Stress Resilience. Neurobiol Stress 2015, 1, 6679,  DOI: 10.1016/j.ynstr.2014.09.004
    406. 406
      Franklin, T. B.; Saab, B. J.; Mansuy, I. M. Neural mechanisms of stress resilience and vulnerability. Neuron 2012, 75, 747761,  DOI: 10.1016/j.neuron.2012.08.016
    407. 407
      Bear, T.; Dalziel, J.; Coad, J.; Roy, N.; Butts, C.; Gopal, P. The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress. Microorganisms 2021, 9, 723,  DOI: 10.3390/microorganisms9040723
    408. 408
      Li, L. F.; Zou, H. W.; Song, B. L.; Wang, Y.; Jiang, Y.; Li, Z. L.; Niu, Q. H.; Liu, Y. J. Increased Lactobacillus Abundance Contributes to Stress Resilience in Mice Exposed to Chronic Social Defeat Stress. Neuroendocrinology 2023, 113, 563,  DOI: 10.1159/000528876
    409. 409
      Dinan, T. G.; Stanton, C.; Cryan, J. F. Psychobiotics: a novel class of psychotropic. Biol. Psychiatry 2013, 74, 720726,  DOI: 10.1016/j.biopsych.2013.05.001
    410. 410
      Chattu, V. K.; Manzar, M. D.; Kumary, S.; Burman, D.; Spence, D. W.; Pandi-Perumal, S. R. The Global Problem of Insufficient Sleep and Its Serious Public Health Implications. Healthcare 2019, 7, 1,  DOI: 10.3390/healthcare7010001
    411. 411
      Irwin, C.; McCartney, D.; Desbrow, B.; Khalesi, S. Effects of probiotics and paraprobiotics on subjective and objective sleep metrics: a systematic review and meta-analysis. Eur. J. Clin. Nutr. 2020, 74, 15361549,  DOI: 10.1038/s41430-020-0656-x
    412. 412
      Smith, R. P.; Easson, C.; Lyle, S. M.; Kapoor, R.; Donnelly, C. P.; Davidson, E. J.; Parikh, E.; Lopez, J. V.; Tartar, J. L. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019, 14, e0222394  DOI: 10.1371/journal.pone.0222394
    413. 413
      Benedict, C.; Vogel, H.; Jonas, W.; Woting, A.; Blaut, M.; Schürmann, A.; Cedernaes, J. Gut microbiota and glucometabolic alterations in response to recurrent partial sleep deprivation in normal-weight young individuals. Molecular Metabolism 2016, 5, 11751186,  DOI: 10.1016/j.molmet.2016.10.003
    414. 414
      Zhang, S. L.; Bai, L.; Goel, N.; Bailey, A.; Jang, C. J.; Bushman, F. D.; Meerlo, P.; Dinges, D. F.; Sehgal, A. Human and rat gut microbiome composition is maintained following sleep restriction. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E1564E1571,  DOI: 10.1073/pnas.1620673114
    415. 415
      Anderson, J. R.; Carroll, I.; Azcarate-Peril, M. A.; Rochette, A. D.; Heinberg, L. J.; Peat, C.; Steffen, K.; Manderino, L. M.; Mitchell, J.; Gunstad, J. A preliminary examination of gut microbiota, sleep, and cognitive flexibility in healthy older adults. Sleep Med. 2017, 38, 104107,  DOI: 10.1016/j.sleep.2017.07.018
    416. 416
      Krueger, J. M.; Opp, M. R. Sleep and Microbes. Int. Rev. Neurobiol. 2016, 131, 207225,  DOI: 10.1016/bs.irn.2016.07.003
    417. 417
      Frey, D. J.; Fleshner, M.; Wright, K. P. The effects of 40 h of total sleep deprivation on inflammatory markers in healthy young adults. Brain, Behavior, and Immunity 2007, 21, 10501057,  DOI: 10.1016/j.bbi.2007.04.003
    418. 418
      Kamada, N.; Seo, S. U.; Chen, G. Y.; Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2013, 13, 321335,  DOI: 10.1038/nri3430
    419. 419
      Yasuda, M.; Kato, S.; Yamanaka, N.; Iimori, M.; Matsumoto, K.; Utsumi, D.; Kitahara, Y.; Amagase, K.; Horie, S.; Takeuchi, K. 5-HT3 receptor antagonists ameliorate 5-fluorouracil-induced intestinal mucositis by suppression of apoptosis in murine intestinal crypt cells. Br. J. Pharmacol. 2013, 168, 13881400,  DOI: 10.1111/bph.12019
    420. 420
      Li, H.-L.; Lu, L.; Wang, X.-S.; Qin, L.-Y.; Wang, P.; Qiu, S.-P.; Wu, H.; Huang, F.; Zhang, B.-B.; Shi, H.-L. Alteration of Gut Microbiota and Inflammatory Cytokine/Chemokine Profiles in 5-Fluorouracil Induced Intestinal Mucositis. Frontiers in Cellular and Infection Microbiology 2017, 7, 455,  DOI: 10.3389/fcimb.2017.00455
    421. 421
      Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355361,  DOI: 10.1038/sj.ejcn.1602546
    422. 422
      Parkar, S. G.; Kalsbeek, A.; Cheeseman, J. F. Potential Role for the Gut Microbiota in Modulating Host Circadian Rhythms and Metabolic Health. Microorganisms 2019, 7, 41,  DOI: 10.3390/microorganisms7020041
    423. 423
      Zhao, D.; Yu, Y.; Shen, Y.; Liu, Q.; Zhao, Z.; Sharma, R.; Reiter, R. J. Melatonin Synthesis and Function: Evolutionary History in Animals and Plants. Front. Endocrinol. 2019, 10, 249,  DOI: 10.3389/fendo.2019.00249
    424. 424
      Wang, B.; Duan, R.; Duan, L. Prevalence of sleep disorder in irritable bowel syndrome: A systematic review with meta-analysis. Saudi J. Gastroenterol. 2018, 24, 141,  DOI: 10.4103/sjg.SJG_603_17
    425. 425
      Miglis, M. G. Autonomic dysfunction in primary sleep disorders. Sleep Med. 2016, 19, 4049,  DOI: 10.1016/j.sleep.2015.10.001
    426. 426
      Tobaldini, E.; Costantino, G.; Solbiati, M.; Cogliati, C.; Kara, T.; Nobili, L.; Montano, N. Sleep, sleep deprivation, autonomic nervous system and cardiovascular diseases. Neurosci Biobehav Rev. 2017, 74, 321329,  DOI: 10.1016/j.neubiorev.2016.07.004
    427. 427
      Vgontzas, A. N.; Chrousos, G. P. Sleep, the hypothalamic-pituitary-adrenal axis, and cytokines: multiple interactions and disturbances in sleep disorders. Endocrinol. Metab. Clin. North Am. 2002, 31, 1536,  DOI: 10.1016/S0889-8529(01)00005-6
    428. 428
      Schlereth, T.; Birklein, F. The sympathetic nervous system and pain. Neuromolecular Med. 2008, 10, 141147,  DOI: 10.1007/s12017-007-8018-6
    429. 429
      Wong, R. K.; Yang, C.; Song, G. H.; Wong, J.; Ho, K. Y. Melatonin regulation as a possible mechanism for probiotic (VSL#3) in irritable bowel syndrome: a randomized double-blinded placebo study. Dig. Dis. Sci. 2015, 60, 186194,  DOI: 10.1007/s10620-014-3299-8
    430. 430
      Zhao, E.; Tait, C.; Minacapelli, C. D.; Catalano, C.; Rustgi, V. K. Circadian Rhythms, the Gut Microbiome, and Metabolic Disorders. Gastro Hep Advances 2022, 1, 93105,  DOI: 10.1016/j.gastha.2021.10.008
    431. 431
      Liang, X.; Bushman, F. D.; FitzGerald, G. A. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 1047910484,  DOI: 10.1073/pnas.1501305112
    432. 432
      Paulose, J. K.; Wright, J. M.; Patel, A. G.; Cassone, V. M. Human Gut Bacteria Are Sensitive to Melatonin and Express Endogenous Circadian Rhythmicity. PLoS One 2016, 11, e0146643  DOI: 10.1371/journal.pone.0146643
    433. 433
      Mukherji, A.; Kobiita, A.; Ye, T.; Chambon, P. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell 2013, 153, 812827,  DOI: 10.1016/j.cell.2013.04.020
    434. 434
      Paschos, G. K.; FitzGerald, G. A. Circadian Clocks and Metabolism: Implications for Microbiome and Aging. Trends Genet. 2017, 33, 760769,  DOI: 10.1016/j.tig.2017.07.010
    435. 435
      Fernandez-Real, J. M.; Serino, M.; Blasco, G.; Puig, J.; Daunis-i-Estadella, J.; Ricart, W.; Burcelin, R.; Fernández-Aranda, F.; Portero-Otin, M. Gut Microbiota Interacts With Brain Microstructure and Function. J. Clin Endocrinol Metab 2015, 100, 45054513,  DOI: 10.1210/jc.2015-3076
    436. 436
      Carlson, A. L.; Xia, K.; Azcarate-Peril, M. A.; Goldman, B. D.; Ahn, M.; Styner, M. A.; Thompson, A. L.; Geng, X.; Gilmore, J. H.; Knickmeyer, R. C. Infant Gut Microbiome Associated With Cognitive Development. Biol. Psychiatry 2018, 83, 148159,  DOI: 10.1016/j.biopsych.2017.06.021
    437. 437
      Polidano, C.; Zhu, A.; Bornstein, J. C. The relation between cesarean birth and child cognitive development. Sci. Rep. 2017, 7, 11483,  DOI: 10.1038/s41598-017-10831-y
    438. 438
      Chung, Y.-C.; Jin, H.-M.; Cui, Y.; Kim, D. S.; Jung, J. M.; Park, J.-I.; Jung, E.-S.; Choi, E.-K.; Chae, S.-W. Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J. Funct. Foods 2014, 10, 465474,  DOI: 10.1016/j.jff.2014.07.007
    439. 439
      Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 13941401.e4,  DOI: 10.1053/j.gastro.2013.02.043
    440. 440
      Smith, A. P.; Sutherland, D.; Hewlett, P. An Investigation of the Acute Effects of Oligofructose-Enriched Inulin on Subjective Wellbeing, Mood and Cognitive Performance. Nutrients 2015, 7, 88878896,  DOI: 10.3390/nu7115441
    441. 441
      Lee, S.-H.; Yoon, S.-H.; Jung, Y.; Kim, N.; Min, U.; Chun, J.; Choi, I. Emotional well-being and gut microbiome profiles by enterotype. Sci. Rep. 2020, 10, 20736,  DOI: 10.1038/s41598-020-77673-z
    442. 442
      Gros, D. F.; Antony, M. M.; McCabe, R. E.; Swinson, R. P. Frequency and severity of the symptoms of irritable bowel syndrome across the anxiety disorders and depression. J. Anxiety Disord. 2009, 23, 290296,  DOI: 10.1016/j.janxdis.2008.08.004
    443. 443
      Mayer, E. A.; Labus, J. S.; Tillisch, K.; Cole, S. W.; Baldi, P. Towards a systems view of IBS. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 592605,  DOI: 10.1038/nrgastro.2015.121
    444. 444
      Zhang, J.; Ma, L.; Chang, L.; Pu, Y.; Qu, Y.; Hashimoto, K. A key role of the subdiaphragmatic vagus nerve in the depression-like phenotype and abnormal composition of gut microbiota in mice after lipopolysaccharide administration. Translational Psychiatry 2020, 10, 186,  DOI: 10.1038/s41398-020-00878-3
    445. 445
      Pu, Y.; Tan, Y.; Qu, Y.; Chang, L.; Wang, S.; Wei, Y.; Wang, X.; Hashimoto, K. A role of the subdiaphragmatic vagus nerve in depression-like phenotypes in mice after fecal microbiota transplantation from Chrna7 knock-out mice with depression-like phenotypes. Brain, Behavior, and Immunity 2021, 94, 318326,  DOI: 10.1016/j.bbi.2020.12.032
    446. 446
      Bruch, J. D. Intestinal infection associated with future onset of an anxiety disorder: Results of a nationally representative study. Brain. Behav. Immun. 2016, 57, 222226,  DOI: 10.1016/j.bbi.2016.05.014
    447. 447
      Kiecolt-Glaser, J. K.; Wilson, S. J.; Shrout, M. R.; Madison, A. A.; Andridge, R.; Peng, J.; Malarkey, W. B.; Bailey, M. T. The gut reaction to couples’ relationship troubles: A route to gut dysbiosis through changes in depressive symptoms. Psychoneuroendocrinology 2021, 125, 105132,  DOI: 10.1016/j.psyneuen.2021.105132
    448. 448
      Kim, H. N.; Yun, Y.; Ryu, S.; Chang, Y.; Kwon, M. J.; Cho, J.; Shin, H.; Kim, H. L. Correlation between gut microbiota and personality in adults: A cross-sectional study. Brain. Behav. Immun. 2018, 69, 374385,  DOI: 10.1016/j.bbi.2017.12.012
    449. 449
      Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E. F.; Wang, J.; Tito, R. Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C. The neuroactive potential of the human gut microbiota in quality of life and depression. Nature Microbiology 2019, 4, 623632,  DOI: 10.1038/s41564-018-0337-x
    450. 450
      Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D. R.; Fernandes, G. R.; Tap, J.; Bruls, T.; Batto, J.-M. Enterotypes of the human gut microbiome. Nature 2011, 473, 174180,  DOI: 10.1038/nature09944
    451. 451
      Costello, E. K.; Lauber, C. L.; Hamady, M.; Fierer, N.; Gordon, J. I.; Knight, R. Bacterial community variation in human body habitats across space and time. Science 2009, 326, 16941697,  DOI: 10.1126/science.1177486
    452. 452
      Wu, G. D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y. Y.; Keilbaugh, S. A.; Bewtra, M.; Knights, D.; Walters, W. A.; Knight, R. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105108,  DOI: 10.1126/science.1208344
    453. 453
      Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M. R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315320,  DOI: 10.1016/j.nut.2015.09.003
    454. 454
      Wallace, C. J. K.; Milev, R. The effects of probiotics on depressive symptoms in humans: a systematic review. Ann. Gen. Psychiatry 2017, 16, 14,  DOI: 10.1186/s12991-017-0138-2
    455. 455
      Le Morvan de Sequeira, C.; Hengstberger, C.; Enck, P.; Mack, I. Effect of Probiotics on Psychiatric Symptoms and Central Nervous System Functions in Human Health and Disease: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 621,  DOI: 10.3390/nu14030621
    456. 456
      Hill, C.; Guarner, F.; Reid, G.; Gibson, G. R.; Merenstein, D. J.; Pot, B.; Morelli, L.; Canani, R. B.; Flint, H. J.; Salminen, S. 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, 506514,  DOI: 10.1038/nrgastro.2014.66
    457. 457
      Dahiya, D.; Nigam, P. S. Clinical Potential of Microbial Strains, Used in Fermentation for Probiotic Food, Beverages and in Synbiotic Supplements, as Psychobiotics for Cognitive Treatment through Gut-Brain Signaling. Microorganisms 2022, 10, 1687,  DOI: 10.3390/microorganisms10091687
    458. 458
      Zawistowska-Rojek, A.; Tyski, S. How to Improve Health with Biological Agents-Narrative Review. Nutrients 2022, 14, 1700,  DOI: 10.3390/nu14091700
    459. 459
      Gibson, G. R.; Hutkins, R.; Sanders, M. E.; Prescott, S. L.; Reimer, R. A.; Salminen, S. J.; Scott, K.; Stanton, C.; Swanson, K. S.; Cani, P. D. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nature Reviews Gastroenterology & Hepatology 2017, 14, 491502,  DOI: 10.1038/nrgastro.2017.75
    460. 460
      Swanson, K. S.; Gibson, G. R.; Hutkins, R.; Reimer, R. A.; Reid, G.; Verbeke, K.; Scott, K. P.; Holscher, H. D.; Azad, M. B.; Delzenne, N. M. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nature Reviews Gastroenterology & Hepatology 2020, 17, 687701,  DOI: 10.1038/s41575-020-0344-2
    461. 461
      Salminen, S.; Collado, M. C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E. M. M.; Sanders, M. E.; Shamir, R.; Swann, J. R.; Szajewska, H. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nature Reviews Gastroenterology & Hepatology 2021, 18, 649667,  DOI: 10.1038/s41575-021-00440-6
    462. 462
      Sarkar, A.; Lehto, S. M.; Harty, S.; Dinan, T. G.; Cryan, J. F.; Burnet, P. W. J. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci. 2016, 39, 763781,  DOI: 10.1016/j.tins.2016.09.002
    463. 463
      Long-Smith, C.; O’Riordan, K. J.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Microbiota-Gut-Brain Axis: New Therapeutic Opportunities. Annu. Rev. Pharmacol Toxicol 2020, 60, 477502,  DOI: 10.1146/annurev-pharmtox-010919-023628
    464. 464
      Pferschy-Wenzig, E. M.; Pausan, M. R.; Ardjomand-Woelkart, K.; Röck, S.; Ammar, R. M.; Kelber, O.; Moissl-Eichinger, C.; Bauer, R. Medicinal Plants and Their Impact on the Gut Microbiome in Mental Health: A Systematic Review. Nutrients 2022, 14, 2111,  DOI: 10.3390/nu14102111
    465. 465
      PitchBook. https://www.pitchbook.com/ (accessed March 30, 2023).
    466. 466
      Black, C. J.; Ford, A. C. Global burden of irritable bowel syndrome: trends, predictions and risk factors. Nature Reviews Gastroenterology & Hepatology 2020, 17, 473486,  DOI: 10.1038/s41575-020-0286-8
    467. 467
      Fecal Microbiota Transplantation. https://www.idsociety.org/public-health/emerging-clinical-issues/emerging-clinical-issues/fecal-microbiota-transplantation/ (accessed November 28, 2022).
    468. 468
      BiomeBank announces world first regulatory approval for donor derived microbiome drug. https://www.biomebank.com/news/media-release/biomebank-announces-world-first-regulatory-approval-for-donor-derived-microbiome-drug/ (accessed November 22, 2022).
    469. 469
      Ferring Receives U.S. FDA Approval for REBYOTA (fecal microbiota, live-jslm) – A Novel First-in-Class Microbiota-Based Live Biotherapeutic. https://ferringusa.com/?press=ferring-receives-u-s-fda-approval-for-rebyota-fecal-microbiota-live-jslm-a-novel-first-in-class-microbiota-based-live-biotherapeutic (accessed December 6, 2022).
    470. 470
      Clinical Trials. https://www.clinicaltrials.gov/ (accessed January 27, 2023).
    471. 471
      Fijan, S. Microorganisms with claimed probiotic properties: an overview of recent literature. Int. J. Environ. Res. Public Health 2014, 11, 47454767,  DOI: 10.3390/ijerph110504745
    472. 472
      Effect of Two Probiotic Formulations on Mental Health and Mood Biomarkers in Adults With Depressive Symptoms. https://clinicaltrials.gov/ct2/show/NCT05564767?term=NCT05564767&draw=2&rank=1 (accessed November 27, 2022).
    473. 473
      Effects of Probiotics on Gut Microbiota, Endocannabinoid and Immune Activation and Symptoms of Fatigue in Dancers. https://clinicaltrials.gov/ct2/show/NCT05567653?term=NCT05567653&draw=2&rank=1 (accessed November 27, 2022).
    474. 474
      Probiotics on Sleep Among Adults Study. https://clinicaltrials.gov/ct2/show/NCT04767997?term=NCT04767997&draw=2&rank=1 (accessed November 27, 2022).
    475. 475
      Rode, J.; Edebol Carlman, H. M. T.; König, J.; Repsilber, D.; Hutchinson, A. N.; Thunberg, P.; Andersson, P.; Persson, J.; Kiselev, A.; Lathrop Stern, L. Probiotic Mixture Containing Lactobacillus helveticus, Bifidobacterium longum and Lactiplantibacillus plantarum Affects Brain Responses Toward an Emotional Task in Healthy Subjects: A Randomized Clinical Trial. Front Nutr 2022, 9, 827182,  DOI: 10.3389/fnut.2022.827182
    476. 476
      Patterson, E.; Griffin, S. M.; Ibarra, A.; Ellsiepen, E.; Hellhammer, J. Lacticaseibacillus paracasei Lpc-37® improves psychological and physiological markers of stress and anxiety in healthy adults: a randomized, double-blind, placebo-controlled and parallel clinical trial (the Sisu study). Neurobiol Stress 2020, 13, 100277,  DOI: 10.1016/j.ynstr.2020.100277
    477. 477
      Stress & Anxiety Dampening Effects of a Probiotic Supplement Compared to Placebo in Healthy Subjects. https://www.clinicaltrials.gov/ct2/show/NCT03494725?term=NCT03494725&draw=2&rank=1 (accessed November 28, 2022).
    478. 478
      The Cognitive Effects of 6 Weeks Administration With a Probiotic. https://www.clinicaltrials.gov/ct2/show/NCT03601559?term=NCT03601559&draw=2&rank=1 (accessed November 28, 2022).
    479. 479
      Probiotic Effects on the Microbe-brain-gut Interaction and Brain Activity During Stress Tasks in Healthy Subjects. https://www.clinicaltrials.gov/ct2/show/NCT03615651?term=NCT03615651&draw=2&rank=1 (accessed November 28, 2022).
    480. 480
      Lactobacillus Plantarum DR7 for Gut-Brain-Axis Benefits (DR7). https://www.clinicaltrials.gov/ct2/show/NCT03370458?term=NCT03370458&draw=2&rank=1 (accessed November 28, 2022).
    481. 481
      Sur, D.; Manna, B.; Niyogi, S. K.; Ramamurthy, T.; Palit, A.; Nomoto, K.; Takahashi, T.; Shima, T.; Tsuji, H.; Kurakawa, T. Role of probiotic in preventing acute diarrhoea in children: a community-based, randomized, double-blind placebo-controlled field trial in an urban slum. Epidemiology and Infection 2011, 139, 919926,  DOI: 10.1017/S0950268810001780
    482. 482
      Cottrell, J.; Koenig, K.; Perfekt, R.; Hofmann, R.; For the Loperamide–Simethicone Acute Diarrhoea Study, T. Comparison of Two Forms of Loperamide–Simeticone and a Probiotic Yeast (Saccharomyces boulardii) in the Treatment of Acute Diarrhoea in Adults: A Randomised Non-Inferiority Clinical Trial. Drugs R&D 2015, 15, 363373,  DOI: 10.1007/s40268-015-0111-y
    483. 483
      Skrzydło-Radomańska, B.; Prozorow-Król, B.; Cichoż-Lach, H.; Majsiak, E.; Bierła, J. B.; Kanarek, E.; Sowińska, A.; Cukrowska, B. The Effectiveness and Safety of Multi-Strain Probiotic Preparation in Patients with Diarrhea-Predominant Irritable Bowel Syndrome: A Randomized Controlled Study. Nutrients 2021, 13, 756,  DOI: 10.3390/nu13030756
    484. 484
      Quigley, E. M. M.; Markinson, L.; Stevenson, A.; Treasure, F. P.; Lacy, B. E. Randomised clinical trial: efficacy and safety of the live biotherapeutic product MRx1234 in patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 2023, 57, 8193,  DOI: 10.1111/apt.17310
    485. 485
      Effect of Lactobacillus Gasseri DSM 27123 on Functional Constipation in Healthy Women. https://www.clinicaltrials.gov/ct2/show/NCT02592200?term=NCT02592200&draw=2&rank=1 (accessed November 28, 2022).
    486. 486
      Dietary Supplementation Effects on Bowel Movement Frequency and Intestinal Biological Markers in Seniors Presenting Slowed Intestinal Transit. https://www.clinicaltrials.gov/ct2/show/NCT04304170?term=NCT04304170&draw=2&rank=1 (accessed November 28, 2022).
    487. 487
      Multi Strain Probiotic Preparation in Patients With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT04662957?term=NCT04662957&draw=2&rank=1 (accessed November 28, 2022).
    488. 488
      The Effect of Probiotic Supported Yogurt Consumption on Gastrointestinal Symptoms. https://www.clinicaltrials.gov/ct2/show/NCT05566171?term=NCT05566171&draw=2&rank=1 (accessed November 28, 2022).
    489. 489
      B. Lactis HN019 for Functional Constipation (CTT). https://www.clinicaltrials.gov/ct2/show/NCT01463293?term=NCT01463293&draw=2&rank=1 (accessed November 28, 2022).
    490. 490
      Effects of Cultura Yoghurt in Relation to Transit Time and Digestive Discomfort in Healthy Women and Men. https://www.clinicaltrials.gov/ct2/show/NCT01102036?term=NCT01102036&draw=2&rank=1 (accessed November 28, 2022).
    491. 491
      A Trial for New Treatment of Adult Participants With Irritable Bowel Syndrome. https://clinicaltrials.gov/ct2/show/NCT03721107?term=NCT03721107&draw=2&rank=1 (accessed Feb 16, 2023).
    492. 492
      Randomized Controlled Field Trial of a Probiotics to Assess Its Role in Preventig Diarrhoea (Yakult). https://www.clinicaltrials.gov/ct2/show/NCT00534170?term=NCT00534170&draw=2&rank=1 (accessed November 28, 2022).
    493. 493
      Probiotics and Hospital Outcome in the Elderly (PROAGE). https://www.clinicaltrials.gov/ct2/show/NCT00794924?term=NCT00794924&draw=2&rank=1 (accessed November 28, 2022).
    494. 494
      Evaluation of the Efficacy of Two Probiotic Strains for Irritable Bowel Syndrome (14PIHL). https://www.clinicaltrials.gov/ct2/show/NCT02213172?term=NCT02213172&draw=2&rank=1 (accessed November 28, 2022).
    495. 495
      A Comparison of Three Medications to Treat Diarrhea in Adults. https://www.clinicaltrials.gov/ct2/show/NCT00807326?term=NCT00807326&draw=2&rank=1 (accessed November 28, 2022).
    496. 496
      A Trial to Evaluate the Effects of Bifidobacterium Longum NCC3001 on Intestinal and Psychological Symptoms in Subjects With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT05054309?term=NCT05054309&draw=2&rank=1 (accessed November 28, 2022).
    497. 497
      Study to Evaluate a Probiotic in Healthy Subjects With a History of Abdominal Discomfort and Bloating. https://www.clinicaltrials.gov/ct2/show/NCT01099696?term=NCT01099696&draw=2&rank=1 (accessed November 28, 2022).
    498. 498
      Efficacy of a Multi-strain Probiotic in the Treatment of Irritable Bowel Syndrome (IBS). https://www.clinicaltrials.gov/ct2/show/NCT01887834?term=NCT01887834&draw=2&rank=1 (accessed November 28, 2022).
    499. 499
      Trial to Evaluate Dietary Supplements to Maintain Gut Health During Travel (P3). https://www.clinicaltrials.gov/ct2/show/NCT04605783?term=NCT04605783&draw=2&rank=1 (accessed November 28, 2022).
    500. 500
      To Study the Efficacy and Safety of L. Plantarum UALp-05TM in Diarrhea- Predominant-irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT04950296?term=NCT04950296&draw=2&rank=1 (accessed November 28, 2022).
    501. 501
      Saccharomyces Cerevisiae for Irritable Bowel Syndrome (IBS). https://www.clinicaltrials.gov/ct2/show/NCT05149599?term=NCT05149599&draw=2&rank=1 (accessed November 28, 2022).
    502. 502
      Johnstone, N.; Milesi, C.; Burn, O.; van den Bogert, B.; Nauta, A.; Hart, K.; Sowden, P.; Burnet, P. W. J.; Cohen Kadosh, K. Anxiolytic effects of a galacto-oligosaccharides prebiotic in healthy females (18–25 years) with corresponding changes in gut bacterial composition. Sci. Rep. 2021, 11, 8302,  DOI: 10.1038/s41598-021-87865-w
    503. 503
      Parilli-Moser, I.; Domínguez-López, I.; Trius-Soler, M.; Castellví, M.; Bosch, B.; Castro-Barquero, S.; Estruch, R.; Hurtado-Barroso, S.; Lamuela-Raventós, R. M. Consumption of peanut products improves memory and stress response in healthy adults from the ARISTOTLE study: A 6-month randomized controlled trial. Clin. Nutr. 2021, 40, 55565567,  DOI: 10.1016/j.clnu.2021.09.020
    504. 504
      Prebiotics and Stress Reduction in Women. https://www.clinicaltrials.gov/ct2/show/NCT05372601?term=NCT05372601&draw=2&rank=1 (accessed November 28, 2022).
    505. 505
      Evaluating the Effects of Prebiotics on Sleep, the Gut Microbiome, Cognition, Immune Function and Stress. https://www.clinicaltrials.gov/ct2/show/NCT05239845?term=NCT05239845&draw=2&rank=1 (accessed November 28, 2022).
    506. 506
      Healthy Prebiotic and Postbiotic Effects of Peanuts and Peanut Butter: College Intervention Trial (ARISTOTLE). https://www.clinicaltrials.gov/ct2/show/NCT04324749?term=NCT04324749&draw=2&rank=1 (accessed November 28, 2022).
    507. 507
      Polyphenols, Prebiotics, the Gut Microbiome and Stress. https://www.clinicaltrials.gov/ct2/show/NCT05528575?term=NCT05528575&draw=2&rank=1 (accessed November 28, 2022).
    508. 508
      Prebiotics and Mental Health: Behavioural. https://www.clinicaltrials.gov/ct2/show/NCT04616937?term=NCT04616937&draw=2&rank=1 (accessed November 28, 2022).
    509. 509
      Selling, J.; Swann, P.; Madsen, L. R., 2nd; Oswald, J. Improvement in Gastroesophageal Reflux Symptoms From a Food-grade Maltosyl-isomaltooligosaccharide Soluble Fiber Supplement: A Case Series. Integr. Med. 2018, 17, 4042
    510. 510
      Ansell, J.; Butts, C. A.; Paturi, G.; Eady, S. L.; Wallace, A. J.; Hedderley, D.; Gearry, R. B. Kiwifruit-derived supplements increase stool frequency in healthy adults: a randomized, double-blind, placebo-controlled study. Nutr. Res. (N.Y.) 2015, 35, 401408,  DOI: 10.1016/j.nutres.2015.04.005
    511. 511
      Blatchford, P.; Stoklosinski, H.; Eady, S.; Wallace, A.; Butts, C.; Gearry, R.; Gibson, G.; Ansell, J. Consumption of kiwifruit capsules increases Faecalibacterium prausnitzii abundance in functionally constipated individuals: a randomised controlled human trial. J. Nutr Sci. 2017, 6, e52,  DOI: 10.1017/jns.2017.52
    512. 512
      SILK, D. B. A.; DAVIS, A.; VULEVIC, J.; TZORTZIS, G.; GIBSON, G. R. Clinical trial: the effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2009, 29, 508518,  DOI: 10.1111/j.1365-2036.2008.03911.x
    513. 513
      Tolerability Study of a Novel Microbiome Therapeutic in Subjects With Gastroesophageal Reflux Disease. https://www.clinicaltrials.gov/ct2/show/NCT04491734?term=NCT04491734&draw=2&rank=1 (accessed November 28, 2022).
    514. 514
      Utility of the Administration of Chesnut and Quebracho Extract for Irritable Bowel Syndrome Diarrhea Predominant. https://www.clinicaltrials.gov/ct2/show/NCT05207618?term=NCT05207618&draw=2&rank=1 (accessed November 28, 2022).
    515. 515
      Evaluation of the Efficacy of a New Specific Infant Formula in Case of Functional Constipation. https://www.clinicaltrials.gov/ct2/show/NCT05340712?term=NCT05340712&draw=2&rank=1 (accessed November 28, 2022).
    516. 516
      ReFerm. https://referm.dk/ (accessed November 27, 2022).
    517. 517
      Effect of Postbiotic Product on Colonic Barriers in IBS. https://www.clinicaltrials.gov/ct2/show/NCT05475314?term=NCT05475314&draw=2&rank=1 (accessed November 28, 2022).
    518. 518
      Pilot Study to Assess the Effect of a Postbiotic Blend on Moderate Self-reported Anxiety (Anx). https://www.clinicaltrials.gov/ct2/show/NCT05562739?term=NCT05562739&draw=2&rank=1 (accessed November 28, 2022).
    519. 519
      Evaluating the Safety and Efficacy of the Probiotic Bifidobacterium Longum ES1 and the Post Biotic Heat-treated Bifidobacterium Longum ES1 (HT-ES1) on IBS Symptom Severity in Patients With Diarrhoea Predominant Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT05339243?term=NCT05339243&draw=2&rank=1 (accessed November 28, 2022).
    520. 520
      König, J.; Brummer, R. J. Faecal microbiota transplantation in IBS ─ new evidence for success?. Nature Reviews Gastroenterology & Hepatology 2020, 17, 199200,  DOI: 10.1038/s41575-020-0282-z
    521. 521
      El-Salhy, M.; Winkel, R.; Casen, C.; Hausken, T.; Gilja, O. H.; Hatlebakk, J. G. Efficacy of Fecal Microbiota Transplantation for Patients With Irritable Bowel Syndrome at 3 Years After Transplantation. Gastroenterology 2022, 163, 982994.e14,  DOI: 10.1053/j.gastro.2022.06.020
    522. 522
      Effects of Faecal Microbiota Transplantation in Patients With IBS. https://www.clinicaltrials.gov/ct2/show/NCT03822299?term=NCT03822299&draw=2&rank=1 (accessed November 28, 2022).
    523. 523
      Fecal Microbiota Transplantation in Patients With Irritable Bowel Syndrome. https://www.clinicaltrials.gov/ct2/show/NCT02092402?term=NCT02092402&draw=2&rank=1 (accessed November 28, 2022).
    524. 524
      RCE With FMT in the Treatment of Childhood Constipation. https://www.clinicaltrials.gov/ct2/show/NCT05035784?term=NCT05035784&draw=2&rank=1 (accessed November 28, 2022).
    525. 525
      FMT Capsules in Treatment of Patients With Insomnia Clinical Research. https://www.clinicaltrials.gov/ct2/show/NCT05427331?term=NCT05427331&draw=2&rank=1 (accessed November 28, 2022).
    526. 526
      Haridy, R. Australia gives world-first regulatory approval to fecal transplant therapy. https://newatlas.com/medical/first-approval-fecal-transplant-gut-microbiome-therapy/ (accessed November 22, 2022).
    527. 527
      Shepherd, T. A coup for poo: why the world’s first faecal transplant approval matters. The Guardian , November 12, 2022. https://www.theguardian.com/australia-news/2022/nov/13/a-coup-for-poo-why-the-worlds-first-faecal-transplant-approval-matters (accessed November 22, 2022).
    528. 528
      FDA Approves First Fecal Microbiota Product. https://www.fda.gov/news-events/press-announcements/fda-approves-first-fecal-microbiota-product (accessed December 2, 2022).
    529. 529
      Shaffer, J. P.; Nothias, L.-F.; Thompson, L. R.; Sanders, J. G.; Salido, R. A.; Couvillion, S. P.; Brejnrod, A. D.; Lejzerowicz, F.; Haiminen, N.; Huang, S. Standardized multi-omics of Earth’s microbiomes reveals microbial and metabolite diversity. Nat. Microbiol. 2022, 7, 2128,  DOI: 10.1038/s41564-022-01266-x
    530. 530
      Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R. B.-Z. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018, 174, 13881405.e21,  DOI: 10.1016/j.cell.2018.08.041
    531. 531
      Zhu, G.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. Probiotics for mild cognitive impairment and Alzheimer’s disease: a systematic review and meta-analysis. Foods 2021, 10, 1672,  DOI: 10.3390/foods10071672
    532. 532
      Białecka-Dębek, A.; Granda, D.; Szmidt, M. K.; Zielińska, D. Gut microbiota, probiotic interventions, and cognitive function in the elderly: a review of current knowledge. Nutrients 2021, 13, 2514,  DOI: 10.3390/nu13082514
    533. 533
      Munawar, N.; Ahsan, K.; Muhammad, K.; Ahmad, A.; Anwar, M. A.; Shah, I.; Al Ameri, A. K.; Al Mughairbi, F. Hidden role of gut microbiome dysbiosis in schizophrenia: Antipsychotics or psychobiotics as therapeutics?. Int. J. Mol. Sci. 2021, 22, 7671,  DOI: 10.3390/ijms22147671
    534. 534
      Cohen Kadosh, K.; Basso, M.; Knytl, P.; Johnstone, N.; Lau, J. Y. F.; Gibson, G. R. Psychobiotic interventions for anxiety in young people: a systematic review and meta-analysis, with youth consultation. Transl Psychiatry 2021, 11, 352,  DOI: 10.1038/s41398-021-01422-7
    535. 535
      Osadchiy, V.; Martin, C. R.; Mayer, E. A. The Gut-Brain Axis and the Microbiome: Mechanisms and Clinical Implications. Clin. Gastroenterol. Hepatol. 2019, 17, 322332,  DOI: 10.1016/j.cgh.2018.10.002
    536. 536
      Cussotto, S.; Strain, C. R.; Fouhy, F.; Strain, R. G.; Peterson, V. L.; Clarke, G.; Stanton, C.; Dinan, T. G.; Cryan, J. F. Differential effects of psychotropic drugs on microbiome composition and gastrointestinal function. Psychopharmacology (Berl.) 2019, 236, 16711685,  DOI: 10.1007/s00213-018-5006-5
  • Supporting Information

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.3c00127.

    • Prebiotic, probiotic, postbiotic, and fecal transplant therapeutic clinical trial data investigating the treatment of mental disorders and DGBI through gut microbiome modulation (XLSX)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect