Open access peer-reviewed chapter

The Therapeutic Potential of the “Yin-Yang” Garden in Our Gut

Written By

Shabarinath Srikumar and Séamus Fanning

Submitted: 10 August 2018 Reviewed: 13 August 2018 Published: 04 December 2018

DOI: 10.5772/intechopen.80881

From the Edited Volume

Role of Microbes in Human Health and Diseases

Edited by Nar Singh Chauhan

Chapter metrics overview

1,144 Chapter Downloads

View Full Metrics

Abstract

The gut microbiota is made up of trillion microorganisms comprising bacteria, archaea, and eukaryota living in an intimate relationship with the host. This is a highly diverse microbial community and is essentially an open ecosystem despite being deeply embedded in the human body. The gut microbiome is continually exposed to allochthonous bacteria that primarily originates from food intake. Comprising more than 1000 bacterial species, the gut microbiota endows so many different functions—so many that can be considered as an endocrine organ of its own. In this book chapter, we summarize the importance of gut microbiota in the development and maintenance of a healthy human body. We first describe how the gut microbiota is formed during the birth of a human baby and how a healthy microflora is established overtime. We also discuss how important it is to maintain the microbiota in its homeostatic condition. A discussion is also given on how alterations in the microbiota are characteristic of many diseased conditions. Recent investigations report that reestablishing a healthy microbiota in a diseased individual using fecal microbial transplant can be used as a therapeutic approach in curing many diseases. We conclude this chapter with a detailed discussion on fecal microbial transplants.

Keywords

  • microbiome
  • microbiota
  • gut
  • antibiotic
  • IBD
  • FMT

1. Introduction

We, animals, live in a microbe-dominated planet. We are all covered, filled, and fueled by bacteria. All body surfaces like the skin, gastrointestinal tract, urogenital, and respiratory tract are in constant contact with the environment and are, therefore, colonized by bacteria. The realities of life associated with a microbe-dominated planet have led to the coevolution of animals with bacteria. This coevolution has led to close inhabitation of bacteria on different surfaces of the human body, especially the gut. Here, many bacteria and their phages, viruses, fungi, archaea, protists, and nematodes intermingle to form a microbial consortia collectively called “microbiome” or “microbiota”. The presence and abundance of each taxonomic group may vary within population based on their access to adequate health care and local sanitation condition or within individuals based on their metabolic, medical, diet, and various other factors.

Despite being embedded deeply in the human body, the gut is essentially an open ecosystem—with constant exposure to environmental factors. The closure of the NIH-funded human microbiome project has given advanced understanding about the composition and functional characteristics of the gut microbiota composition. The composition of the human microbiome varies significantly depending on the habitat [1]. For example, the gut microbiota is mainly populated by bacteria [2], while the skin harbors mainly fungi [3]. It was always considered that human microbiome outnumbered human nucleated cells by at least a factor of 10. However, reports suggest that the ratio is closer to 1 [4]. Recently metagenomic analysis concluded that the gene set of the human microbiome is 150 times larger than the human gene complement raising the possibility that the large bacterial genetic repertoire aids the human component in performing the essential functions that are not encoded by the human genome. Since more than 99% of these genes are bacterial in origin, the number of bacterial species were calculated to be around 1000–1150 bacterial species. The figures collectively emphasize the biological importance of the microbiome, and the genetic complement can be rightly considered as the second genome [2]. However, the taxonomic diversity of the gut microbiome notwithstanding, this chapter will deal only with the bacterial populations and all further references to gut microbiota means gut bacterial microbiota.

A typical human gut microbiota contains about 1014 bacteria and is made up of more than 1000 bacterial species [5, 6]. The human gut microbiota is composed of six bacterial phyla—Firmicutes, Proteobacteria, Bacteroides, Fusobacteria, Actinobacteria, and Verrucomicrobia. Of this, Bacteroides and Firmicutes occupy 70–90% of the total bacteria present in a healthy gut, while others are present in lower abundances [7]. Under healthy circumstances Proteobacteria and Verrucomicrobia members are also present but in lesser abundance [8]. The huge complexity and the variability of the microbiome make the determination of precise metabolic functions and the host-microbe cross talk very difficult. However, recent advances in deep sequencing and computational biology have contributed to large advances into understanding the unique biology of the gut microbiota and the subject is still in its infancy.

The functions bestowed by the bacterial gut flora are so enormous that it can be considered as an endocrine organ on its own [9]. It is well understood that the gut microbiota and their metabolites play important roles in host homeostasis, such as providing important nutrients like secondary bile salts and B/K group vitamins [10, 11], help in fermenting the otherwise indigestible complex plant carbohydrates such as dietary fibers into short-chain fatty acids (SCFA) [12], contributing to an effective intestinal epithelial barrier and activation of both innate/adaptive immune responses of the host [13]. In addition, the healthy gut microbiome drives intestinal development by promoting vascularization, villus thickening, mucosal surface widening, mucus production, cellular proliferation, and maintaining epithelial junctions [14, 15, 16]. The influence of the gut microbiota, either directly or indirectly, affects the physiology of most host organs even the brain [14, 17, 18, 19, 20].

The taxonomic composition of the microbiota is very subtle—subject to change with variations in the diet [21], feeding time changes [22], sleep wake cycles, and even jet lag [23]. The unique taxonomic signature of the gut microbiota has to be strictly maintained for a healthy gut. Any disruption of the taxonomic composition of the gut could lead to conditions such as inflammatory bowel syndrome (IBS), asthma, obesity, metabolic syndrome, and cancer [24]. In some cases, resuscitation of the gut microbiota using probiotic bacteria like Bifidobacteria and Lactobacillus leads to the mitigation of gut inflammation consequently regaining health.

In this chapter, we will sequentially detail how the microbiota establishes itself in a human body, how minor or major variations in the gut microbiome introduces diseased conditions, and how the recent therapeutic approaches aimed at resuscitating the healthy microbiota can cure many dysbiotic microbiota-associated conditions.

Advertisement

2. Our internal garden—on how gut microbiota is planted and nurtured

How do we become colonized with gut microbiota? Where do we get our initial inoculum from and how does this initial inoculum proceed in to a well-flourished, well-established microbiota? The first step to understanding this is to identify the initial inoculum and how this inoculum develops into a the full-fledged adult ecosystem following a series of ecological succession steps.

2.1 The initial colonization

The most prevalent concept, and a very incorrect one, is that babies are borne sterile and after birth, the body becomes immediately colonized with microbes from the surrounding environment. Placental mammals such as humans are borne through a birth canal, which is colonized by microbes. A baby acquires its first inoculum from the birth canal. A healthy vaginal microbiota is composed of few bacterial species [25, 26] and is predominated by Lactobacilli [27]. Consequently, naturally borne babies acquire vaginal microbes like Lactobacillus, Prevotella, and Sneathia spp. [28]. The bacteria are present in the mouth, skin, and even in the meconium. Therefore, all neonates are colonized by essentially the same vaginally derived bacteria obtained vertically from the mother. Once this microbiota is established, the microbiota becomes highly differentiated depending on their ability to colonize different body sites. For example, in the gut, facultative anaerobes establish and reduce the environment [29]. This highly reduced environment facilitates the colonization of obligate anaerobes [30, 31, 32, 33]. In addition, breastfeeding will also enrich vaginally acquired lactic-acid-producing bacteria in the baby’s intestine [34]. From then on, it is the physiology of the host habitat that selects the community that becomes well adapted to colonize that particular habitat. For example, the physiochemical, immunological, and the diet play an important role in determining the microbial community that will colonize the small and large intestines. The host genotype also appeared to influence the composition of the gut microbiota [35].

Cesarean section babies, in contrast to vaginally borne babies, are dominated by skin-associated bacteria like Staphylococcus, Corynebacterium, and Propionibacterium spp. [28]. The Staphylococcal-rich microbiota could be obtained from the skin of the humans the baby is in contact with. The lack of a natural first inoculum in C-section babies affects the bacterial community in the GI tract [36, 37]. This variation from the naturally borne baby’s microbiome will increase the susceptibility of the C-section babies to certain pathogens. For example, about 70% of the MRSA-caused skin infection happens to C-section babies. In addition, there is an additional risk to atopic diseases [38], allergies, and asthma [28, 39].

2.2 Development of the microbiota

The establishment, composition, and the density of the microbiome in the gastrointestinal (GI) tract depends on the biochemical factors like pH, oxygen gradient, antimicrobial peptides (AMPs), bile salts, etc. There is a pH gradient across the GI tract—lowest in the stomach and gradually increase to the terminal ileum, and a drop in caecum and increases toward the distal colon. Oxygen also exhibits a gradient across the length of the tract. The levels are highest in the upper GI tract, which decrease to anaerobic conditions in the distal colon. Radially across the tract too, there exists a oxygen gradient. Anoxic conditions exist in the lumen, while there is an increase in the oxygen tension near the mucosa, and this oxygen is rapidly consumed by the facultative anaerobes [40]. Each area in the gut produces its own AMPs. Saliva contains lysozyme, which is antibacterial. Small intestine produces α-defensins, C-type lectins, lysozyme, and phospholipase A2. The large intestine produces β-defensins, C-type lectins, cathelicidins, galectins, and lipocalin. Mucus also plays an important role in the distribution of the gut microbiota. The mucus in the stomach and colon can be discriminated into two layers—outer loose layer, which is densely populated by bacteria, and the inner “solid” layer, where bacteria is sparce [41, 42]. Only mucin-degrading bacteria like Akkermansia muciniphila reach to the inner solid layer. Some pathogens like Helicobacter pylori, Salmonella, Yersinia, Campylobacter, etc. can also reach the inner mucus layer [43, 44]. All these factors play an important role in the establishment and the distribution of the gut microbiota.

The initially colonized microbiome has relatively few bacterial species. But during the initial phase of life, bacterial diversity increases in the microbiota. This may be because of the constant exposure of the baby to the environment. The gradual increase in the length of the GI tract can also provide a new niche for the bacteria to colonize. The bacterial diversity also depends on the diet as the introduction of a more plant-based diet increases the proportion of the Firmicutes [45]. Though lifestyle, illness, puberty, and other variable factors affect the microbiome, family members tend to have similar microbiomes with shared bacterial strains [35]. Thus, during the first year of life, the microbiome proceeds through a very variable phase. A distinct composition resembling the “adult microbiome composition” is established once an adult diet is established after weaning [35]. Once established, the gut microbiome composition seems to remain stable for a long time, possibly lifelong [46].

Advertisement

3. Maintain the flora!—on how any alterations could be disastrous

Biologically, the gut microbiome is very essential for the normal functioning of the human body. This complex ecosystem is responsible for many critical functions like (1) metabolism and energy regulation [47]—up to 10% of our daily consumed calories are provided by the microbes who break down complex plant-derived carbohydrates into short-chain fatty acids (SCFA), the main energy source of the enterocytes. From this perspective, alterations in the gut microbiome can contribute to obesity [48, 49] and consequently type II diabetes [50]; (2) immune system activation [51, 52, 53]—the colonic mucosal immune system plays a dual role and in that it must tolerate the gut microbiome and at the same time react against pathogenic organisms. This homeostasis is achieved by the intricate interplay between the microbiome and the host; (3) colonization resistance—physiologically colonized body surfaces are intrinsically protected from pathogen colonization. It is the intricate interplay between the above mentioned three major functions of the gut microbiota that brings about the physiological healthy state of the host.

Colonization resistance is the native ability of the host to suppress the invasion by exogenous microorganisms [54]. The concept of colonization resistance originated from the studies of Dubos in 1965 who demonstrated that indigenous gut microbiota neutralize colonization by a potential pathogen [55]. Slightly earlier, it was noted that loss of obligate anaerobic bacterial population in the lower intestinal tract correlated with infection, suggesting that the commensal anaerobic organisms were providing colonization resistance [56, 57, 58, 59]. At the time, colonization resistance was thought to result from microbe-mediated inhibition. We now know that the multiple mechanisms like microbiota-mediated activation of host immune responses are also involved. Colonization resistance provides broad protection against bacteria, virus, and other categories of pathogens [60]. On the other hand pathogenic microorganisms can out compete commensal microorganisms, subvert the immune response and invade the epithelia. For example, some pathogens can cause inflammation in the gut and utilize the consequent nutrient-rich inflammatory environment to outgrow other Proteobacteria. However, in a healthy intestine, the gut microbiota maintains the stiff colonization resistance by three mechanisms (1) directly inhibiting or killing the invading organism, (2) maintaining a protective musical barrier, and (3) stimulating a strong immune response that can neutralize a pathogen.

3.1 Direct inhibition

Bacteria produce many bioactive molecules, such as antimicrobial peptides, bacteriocins, etc., to selectively kill or inhibit the growth of competing bacteria [61]. These bioactive molecules are the primary source of antibiotics in the pharmaceutical industry [62].

3.2 Barrier maintenance

Gut microbiota regulates the strength of the intestinal barrier and sequesters themselves within the intestine. For example, the mucus layer is an important deterrent for many pathogenic microorganisms to reach the underlying epithelial cells. Mucus production is enhanced when a germ-free mice epithelium is exposed to some bacterial products [63], which means that an intact microbiota is essential to maintain the required thickness of the mucosal layer to keep pathogenic microorganism at bay. Diet can also influence the thickness of the mucosal layer. An assessment of intestinal microbiota localization with immunofluorescence shows that the absence of microbiota-accessible carbohydrates in the diet resulted in a thinner mucosal layer, thus exposing the underlying epithelial cells to pathogenic organisms [64]. The thinning of the mucosal layer made mouse susceptible to colitis.

3.3 Immune maturation and inflammation

A healthy microbiota is essential for a healthy immune system. Almost one and half decades ago, it was observed that microbial products secreted from the microbiota induced the colonic immune system by activating anti-inflammatory cells and cytokines. In 2005, a polysaccharide from Bacteroides fragilis, an important member of the gut microbiota, was shown to be important in the cellular and physical maturation of a developing immune system [51]. Perhaps, the most immunologically characterized bacterial metabolite synthesized by the microbiota is the Short Chain Fatty Acid (SCFA). In 2009, it was shown that SCFA directly bind G-protein-coupled receptor (GPR43) and activated immune responses [65]. Butyrate is the most characterized SCFA. Butyrate was shown to induce the differentiation of colonic regulatory T cells (Treg) cells in mice. A comparative NMR-based metabolome analysis showed that the concentration luminal butyrate correlated with the number of Treg cells in the colon [52]. Treg cells expressing transcription factor Foxp3 are also important in regulating intestinal inflammation. It was also found that SCFA plays an important role in regulating the function and the size of the colonic Treg pool [53].

Microbes can also directly activate the colonic immune system. Segmented filamentous bacterium (SFB) from the microbiota was shown to adhere tightly to epithelial cells of the terminal ileum with Th17 cells, and this adherence correlated with the induction of inflammatory/antimicrobial defense genes [66]. More recently, bacteria in human feces were subjected to selection based on their potential to induce anti-inflammatory T regulatory cells. It was found that bacteria belonging to cluster XIVa clostridial group induced anti-inflammatory T regulatory cells along with bacteroides species [67, 68]. Gut microbiota can also activate the expression of bacterial C-type lectins in intestinal epithelial cells. The lectin, RegIIIγ, is essential to create a 50-μm clearance zone between the gut microbiota and small intestinal epithelial cells. Abrogation of the RegIIIγ synthesis increased the proximity of gut microbiota to the epithelial cells [69]. This shows that microbiota-activated lectin synthesis can directly act to suppress bacterial activity. In addition, gut microbiota can also enhance systemic antiviral activity [70]. Therefore, it is very important to maintain a healthy microbiota to drive efficient pro-inflammatory and anti-inflammatory immune responses in the host.

Simple alterations to the gut microbiota can often lead to very unhealthy consequences. For example, in the esophagus, the composition of the microbiome is heavily dependent on the microbes originating from the oral cavity and is dominated by Streptococcus, Prevotella, Veillonella, and Fusobacterium [71, 72, 73]. Any alteration to the microbiota composition could lead to inflammation and tumorigenesis. Such altered microbiota compositions were consistent with conditions like gastro-esophageal reflux disease (GERD), Barrett’s esophagus (BE), and adenocarcinoma of the gastro-esophageal (GE) junction. Here, Streptococcus were found to be depleted while Veillonella, Prevotella, Campylobacter, Fusobacterium, Haemophilus, and Neisseria were enriched [74, 75]. Certain taxa present in the oral cavity like Campylobacter concisus and Campylobacter rectus were found to be enriched in the diseased mucosa-associated with GERD and BE [76, 77]. Similarly, for eosinophilic esophagitis (EoE), increased levels of Neisseria, Corynebacterium, and Haemophilus are reported [78].

The most important stomach associated bacterium is H. pylori. H. pylori has symbiotically co-evolved with humans and therefore are highly adapted to humans [79]. Early life time infection of H. pylori is beneficial for humans because it significantly lowers the risk of asthma in later years [80]. This beneficial association is brought about by the immune system modulation by the bacterium due to the high induction of regulatory T cells. The bacterium therefore qualifies for the position of a pathobiont -host determines whether the bacteria remains as a harmless symbiont or becomes pathogenic in nature. In a diseased condition of the host, the bacterium outcompetes the normal microbiota in numbers and becomes the most dominant pathogen.

Sampling the fecal material represents the colonic microbial population. However, sampling the small intestine is difficult because it is accessible only by invasive sampling. The small intestine is populated by distinct microbial communities that are less diverse and are dominated by Veillonella, Streptococcus, Lactobacillus, and Clostridium [81, 82, 83]. In the small intestine, alterations in the microbiome are associated with celiac disease (CeD). Gut microbiota is able to differentially degrade gluten. In CeD patients, there is an over growth of an opportunistic pathogen Pseudomonas aeruginosa producing a elastase called LasB. This enzyme degrades gluten and releases peptides that translocate the intestinal barrier, triggering a T-cell response [84]. A small-intestine-associated autoimmune disease where microbiome plays an important role is graft versus host disease (GvHD)—caused by the activation of T cells where host cells are recognized as antigens cause autoimmune attacks in the GI tract, liver, lung, and skin [85]. Germ-free mice had less propensity to develop GvHD—this led to the thought that microbiome could play an important role [86, 87, 88]. Loss of microbiome diversity and consequent butyrate deprivation pushed cells to apoptosis bearing hallmark histological signs associated with GvHD. An overabundance of Enterococcus (E. faecium and E. faecalis) was observed in patients with GvHD associated with hematopoietic stem cell transfer confirming the association of GvHD with alteration in the microbiome diversity [89].

3.4 Antibiotic-associated colitis

Antibiotics considered as “wonder drugs” were implemented into therapy years before and have saved millions of lives. Even though antibiotics can reduce morbidity and mortality associated with bacterial illness, no antibiotic is pathogen selective. The application of antibiotics lead to collateral damage of accompanying microorganisms in a population, for example in a microbiota. Studies investigating the impact of antibiotics on microbiota confirm that antibiotic treatment increases the susceptibility of an individual to bacterial pathogens by compromising colonization resistance [90, 91, 92, 93].

The observation that antibiotic therapy reduced colonization resistance making the host susceptible to bacterial infections was observed very early in the literature [56, 57, 58, 59]. Gut microbial compositional analysis of an antibiotic-treated mice showed the expansion of γ-proteobacteria and enterococci, suggesting that gut microbiota somehow suppressed the expansion of oxygen-tolerant species [94, 95]. A study on healthy volunteers treated for a week or less with antibiotics reported persistent effects on their bacterial flora that included a loss of biodiversity on the gut flora, insurgence of antibiotic resistance strains, and upregulation of antibiotic resistance genes [96]. Antibiotic treatment can also induce long-term defects in the microbiota. For example, a single dose of clindamycin induced long-term susceptibility to Clostridium difficile infection [92]. A prior treatment with antibiotics not only disturbed the gut microbiota enabling the expansion of pathogenic commensals but also helped exogenic bacterial pathogens to establish inside the gut. When antibiotic-treated mice was infected with vancomycin-resistant Enterococci (VRE), the bacteria displaced the whole normal microbiota of the small and large intestine. In the clinical setting, this initial domination by the VRE preceded the bloodstream infections in patients undergoing hematopoietic stem cell transplant [91].

Microbiota establishes itself very early in the life cycle of every human, and this development is very crucial for a healthy lifestyle [97]. So, administration of antibiotics in the early stages of life predisposes the individual to diseases in late infancy or adulthood, particularly allergic or metabolic syndromes [28]. Antibiotic exposed prenatal mice resulted in exacerbated asthma following intranasal challenge with ovalbumin [98]. This case is true in children who are administered with antibiotics in the first year of life and may develop asthma during sixth or the seventh year [99]. Early use of marcolids in Finnish children led to the development of asthma and increased BMI associated with a dysbiotic gut microbiota [100]. Effects of antibiotic administration in early life are not limited to development of asthma alone but also to obesity. A low dose of penicillin delivered at birth transiently shifted the microbiota, and this transient shift induced sustained effects in body composition, leading to obesity [101]. All these reports emphasize the detrimental microbiota-associated effects of antibiotics and their implications in health.

Clostridium difficile is perhaps the most characterized pathogen associated with antibiotic associated colitis [102]. With >25,000 annual cases worldwide, C. difficile colitis is almost always associated with prior antibiotic use. The suspicion that microbiota-mediated colonization resistance resisted C. difficile in the gut was finally proven in 2013 [103]. 16S rDNA sequencing alone could distinguish between C. difficile-associated diarrhea and C. difficile-negative diarrhea [104]. Antibiotic treatment reduces secondary bile salt production making the host susceptible to C. difficile infection [105, 106]. Apart from C. difficile, Klebsiella oxytoca also caused antibiotic-associated hemorrhagic colitis (AAHC)—a patchy hemorrhagic colitis usually observed after penicillin therapy typically dominating the right colon. Here, the pathogen is intrinsically resistant to β-lactams and the production of enterotoxin tilivalline can lead to intestinal epithelial apoptosis and colitis [107, 108]. Antibiotic-treated mice had impaired innate and adaptive antiviral immune response, and when the mucosa was exposed to influenza virus, the clearance was substantially delayed. On the other hand, these mice had severe bronchiole epithelial degeneration and increased host mortality when exposed to influenza. This is due to the macrophages from an antibiotic-treated mice had decreased expression of genes associated with antiviral immunity [70].

3.5 Inflammatory bowel disease (IBD)

IBD perhaps is the first diseased condition where alterations in microbiota are studied most extensively. IBD is a term mainly used to describe two disease conditions—Crohn’s disease and ulcerative colitis. Here, intestinal cells play an important role in integrating the interactions among intestinal microbiota, mucosal immune system, and environmental factors [109]. It was observed very early that IBD conditions had a genetic component—there was a 10-fold increase in risk if related closely to the patient [110]. Genome-wide association studies (GWAS) reported many genetic factors that are associated with IBD [111]. As more GWAS based studies began identifying genetic factors associated with IBD, it was soon noted that some IBD genetic factors were also associated with other disease conditions like diabetes [112]. Curiously, GWAS and meta-analysis identified considerable overlap between susceptibility loci for IBD and mycobacterial infections [113]. The genetic associations notwithstanding, alterations in the gut microbiome of IBD patients have always been an interesting topic for microbiome researchers. The most significant alteration in the composition of the gut microbiome associated with IBD is the reduction in the abundance of the protective bacterium Faecalibacterium prausnitzii [114]. However, patterns of gut microbiota dysbiosis was not consistent across different studies. In a large cohort study involving more than 400 pediatric cases, multiple samples obtained from multiple locations of the GI tract before and after the onset of Crohn’s disease were analyzed. Increased abundance of Enterobacteriaceae, Pasteurellaceae, Veillonellaceae, and Fusobacteriaceae and decreased abundance of Erysipelotrichales, Bacteroidales, and Clostridiales were found to be strongly consistent with the diseased condition. Oddly enough, there seems to be prevalence of oral bacteria in IBD and Crohn’s disease patients. For example, the prevalence of oral and stomach-associated C. concisus was very high in both diseased conditions [115]. Furthermore, another Gram-negative oral bacteria Fusobacterium nucleatum was found to be abundant in Crohn’s disease [116]. F. nucleatum was shown to be highly proinflammatory and protumorigenic [117, 118, 119]. The bacterium can activate the epithelial cell proliferation and induce a protomeric microenvironment, while inactivating the immunological tumor surveillance.

3.6 Colorectal cancers (CRC)

CRC is the fourth leading causes of death causing cancer and is the third important cause of malignancy. The CRC incidence is growing fast worldwide in low and middle east countries and is expected to increase by 60% by 2030 worldwide [120]. The transformation from a healthy epithelial cell to a malignant cell requires three steps: (1) induction of oncogenic mutations within Lgr5+ intestinal stem cells, (2) altered β-catenin/Wnt signaling, and (3) proinflammatory cascades such as TNFα-NFκB and IL16-STAT3 catalyzing CRC development (Garret 2015 EN). Initially, there was increasing evidence about the role of bacteria in CRC. Bacteria such as Fusobacterium nucleatum, E. coli, and Bacteroides fragilis were shown to be associated with CRC [117, 121]. F. nucleatum was first shown to be highly enriched in tumors [117, 122]. The bacterium produces the FadA antigen, a ligand of E-cadherin in the intestinal epithelial cells that activate the β-catenin pathway leading to uncontrolled cell growth [119]. Furthermore, F. nucleatum is shown to be overrepresented in the colonic mucosa in the cases where the CRC relapses postchemotherapy. This was shown to be an interplay of intricate mechanisms including TLRs, miRNAs, and autophagy induction [123]. Some strains of E. coli harbor the polyketide synthase (pks) island encoding colibactin, capable of inducing DNA damage and mutation in epithelial cells [121]. A metagenome-wide association study on stools collected from patients with advanced adenomas, CRC, and healthy controls identified that certain Bacteroides species (B. dorei, B. vulgaris, B. massiliensis) and E. coli were overrepresented in the microbiome. Similarly, Parvimonas, Bilophila wadsworthia, Fusobacterium nucleatum, and Alistipes spp. were also overrepresented, suggesting that the gut microbiome signature can be used for early diagnosis and treatment [124].

Advertisement

4. The therapeutic potential of the gut microbiota

Humans have used live bacteria, particularly probiotic bacteria, for therapeutic purposes from time immemorial. We have seen some examples in earlier sections of this chapter. Perhaps, the best example of using live bacteria to cure infectious disease comes from antibiotic-associated CDI illness. Clostridium scindens, an obligate anaerobic bacterial species that inhabits the colon, has the rare ability to convert primary bile salts to secondary bile salts and is highly associated with resistance to C. difficile colitis [125]. Administration of C. scindens to susceptible mice resuscitated the secondary bile salt deficiency and rendered the animal more resistant to CDI. C. scindens and C. difficile have a negative correlation-could be the reason for C. difficile resistance in a healthy human gut microbiota [105]. However, the clinical benefit of using a single bacteria is limited. This is because, as we have seen earlier, many of the disorders are caused by a dysbiotic microbiota. Since a microbiota is very diverse in nature, resuscitation of a healthy gut microbiota cannot be achieved by the administration of a single bacterium. The concept of “putting back the bugs” was demonstrated in 1993 by using a combination of probiotic strains to cure chronic constipations and IBS [126]. Even with CDI, a cocktail of 10 gut commensal bacteria including obligate anaerobes could effect a cure [127]. Since then, many experiments have shown that by replenishing the healthy composition of a normal microbiota, many disease conditions can be controlled. Therefore targeting gut microbiota has gathered much attention and many options are currently being evaluated to achieve this goal of re-establishing the healthy gut microbiota to regain health—leading to the concept of fecal microbiota transplant (FMT).

FMT is the procedure where fecal matter is collected from a tested donor, diluted in an isotonic solution, strained, and transplanted into the patient using colonoscopy, endoscopy, sigmoidoscopy, or enema. The history of using stool of healthy donors to treat human diseases dates back to the fourth century in China, during the Dong Jin dynasty (AD 300–400 years) [128]. Fecal suspensions or “yellow soup” was used to treat serious disorders such as food poisoning, febrile disease, typhoid fever, etc., becoming the first record of the utilization of human feces to treat human diseases. There are striking similarities between the earlier yellow soup and modern day FMT technology—(1) the inoculum originated from human fecal matter, (2) administration route is digestive tract, and (3) the fecal matter re-establishes the microbiota thereby treating the disease. This long tradition might be the reason why FMT is so well accepted in China [129]. In Europe, the first report of using fecal enema to treat pseudomembranous colitis came in 1958 [130]. Currently, FMT stands in the threshold of becoming a great technology to cure many disorders considered incurable in the past.

4.1 In treating diseases associated with gut microbiota-associated dysbiosis

CDI perhaps was the first condition in which a treatment was attempted using FMT. In 1983, it was shown that by re-establishing the healthy gut microbiota using FMT, mitigation of CDI can be achieved (Schwan 1983 EN). Severe CDI cases can lead to intensive care admission, sepsis, toxic megacolon, and can prove fatal. Colectomy is the standard method of treatment, but the mortality rate is 50%. In a study involving 29 patients who underwent FMT plus vancomycin for severe CDI cases, 62% of the patients were cured in a single FMT, while 38% needed multiple FMTs. Taken together, FMT was highly efficient for CDI infections [131]. The primary and secondary cure rates with FMT using fresh fecal sample to cure CDI is 91 and 98% [132]. FMT from frozen fecal sample also gave similar efficacies in treating CDI [133, 134]. By 2013, FMT was made officially the treatment strategy for CDI [103, 135]. Many pharmaceutical firms are actively working to bring easily consumable CDI-targeted drugs based on FMT. A defined microbial ecosystem therapeutics (MET-1 or RePOOPulate) was developed to cure recurrent CDI [136, 137]. The closest enema-based drug that is awaiting clinical approval is RBX2660, which depends upon the microbial suspension provided from the donor and is formulated for therapeutic delivery. With positive results in phase 2, the drug is currently in phase 3.

Similar to CDI, IBD is also a dysbiotic-associated disease where FMT is a potential therapy. However, in IBD, the use of FMT is a bit complicated and less efficient than in CDI. Early studies using FMT to treat IBD showed very promising result with good microbiota remission reported over long-term follow up [138]. With years, the outcomes started to differ depending upon sample size, treatment approaches, and study designs [139]. Even within IBD patients, remission rates were different. Crohn’s disease had a higher remission of 61%, while ulcerative colitis patients had a remission rate of 22%. It is clear that the FMT treatment for IBD is complicated by numerous factors like differences in treatment regimens, stool preparation/formulation, and dosing frequencies. Varied levels of dysbiosis and difference in the microbiota composition between donor and patient also complicate FMT treatments. However, it was reported that FMT with intensive doses and multiple donors induced clinical remission and endoscopic improvement in ulcerative colitis patients, and this treatment had distinctive improvements in the microbiota composition [140]. It was also shown that a second FMT 3 months past the first one greatly improved the efficacy and safety in treating IBD with FMT [141, 142]. Here, the patients received FMT repeatedly in 3 month intervals—in a procedure called step-up FMT. The efficacy of the procedure increased at each step and was best suggested for patients with refractory IBD and immune-related diseases [143, 144]. There are currently 27 ongoing clinical trials using FMT targeting IBD with two additional trials on children with IBD [145].

Cancers like colorectal cancers that are associated with a dysbiotic microbiome opening the possibility for a therapeutic intervention using FMT. It was shown that bacteria like Enterococcus hirae and Barnesiella intestinihominis strengthen cyclophosphamide-induced therapeutic immunomodulatory effects in cancer [146]. This has been highlighted very recently with evidence that microbiome influences with body’s ability to respond to antibody therapy for cancers [147, 148]. A correlation was observed between commensal microbial composition and clinical response to anti-PD-L1 therapy through abundance of bacterial species like Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium [149]. When fecal matter from responding patients were transplanted to germ-free mice, the animals were noted with stronger tumor control, augmented T cell responses, and better efficacy [150]. Gut microbiota could also affect anticancer responses with CTLA-4 [151]. The effects of radiation in gut microbiota and the clinical implications of a modified microbial balance postradiotherapy are now being investigated [152]. The microbiota can be modified to improve its efficacy and reduce the toxic burden of these treatments [153]. FMT can be used to reduce the radiation-induced toxicity and the increase the survival rate in irradiated mice. Here, the WBCs, GI tract function, and intestinal epithelial cell integrity were improved [154]. The research advances notwithstanding, therapeutic approaches associated with FMT is still in its nascent stage. However, considerable progress made in this area of research indicate that the application of FMT based therapy to mitigate mortality associated with diseases like cancer is a near possibility.

FMT is also showing great promise in patients undergoing hematopoietic stem cell transplantation surgery. Administration of a series of prophylactic antibiotics during surgery can result in the loss of microbial gut diversity and antibiotic resistant strains like Streptococcus viridans, Enterococcus faecium, and other Enterobacteriaceae can expand their population in the gut. This loss of microbial diversity during stem cell transplantation is associated with marked increase in mortality [91, 155, 156]. Restoration of a healthy microbiota by eliminating the dominant pathogenic microorganisms therefore becomes very important strategy from a therapeutic point of view. FMT involving a consortium of obligate anaerobic commensal bacteria containing especially Barnesiella is shown to eliminate E. faecium in mice [157], opening up a new therapeutic approach for stem cell transplant patients.

4.2 Collection, preparation, and delivery of FMT samples

Collection and preservation of the stool samples carry the primary importance in FMT. Freshly collected feces can either be immediately used, lyophilized, or cryopreserved. The efficacy of FMT in treating CDI using fresh or frozen feces varied but not significantly [158]. The cure rate was 100% in patients receiving fresh feces, 83% for the lyophilized group, and 78% for patients receiving frozen feces. But the efficacy was more pronounced in treating IBD, and this was demonstrated to be caused due to loss of bacteria in frozen feces [143]. The laboratory preparation methods of FMT is also critical for the success of FMT. Recent studies have reported that some preparation methods can stress the living microbial cells affecting the efficacy of FMT [159] emphasizing the need for extreme care. For example, Faecalibacterium prausnitzii is affected when the fecal sample is exposed to oxygen. Currently, the preparation methods can be classified into “rough filtration” (RF), “filtration plus centrifugation” (FPC), and “microfiltration plus centrifugation” (MPC) [141, 142]. Manual preparation methods takes about 6 hours to complete [160]. With the introduction of automated systems and close cooperation between laboratory scientists and clinicians, the time period of preparation from “defecation to freezing” has been shortened to 1 hour [160]—has effectively increased the efficacy of FMT when tested against IBD patients [143]. Current FMT delivery technologies include delivery of the microbiota to upper, mid, and the lower gut [161]. Oral intake of capsular microbiota delivers it to the upper gut [134, 162]. A suspension of microbiota infusion can be transferred to the small intestine beyond the second duodenal segment through endoscopy [163], nasojejunal tube [143], mid-gut transendoscopic enteral tubing (TET) [164], and percutaneous endoscopic gastro-jejunostomy (PEG-J) [161]. The TET procedure for microbiota transplant is considered very successful [164]. Delivery of microbiota to the lower gut could be through colonoscopy, enema, distal ileum stoma, colostomy, and colonic TET [161]. Colonic TET is recommended for patients needing frequent FMT.

Several groups are developing stool products that can be packaged, transported, commercialized, and easily administered by physicians or consumed by patients. These products range from basic (frozen or freeze-dried stool) to more advanced products like capsules of synthetic stool grown in culture and assembled. The most basic products, from stool banks like OpenBiome and Advancing Bio, provide hospitals with screened frozen material ready for clinical use. More advanced are products like RBX2660, a cryopreserved filtered microbiota derived from stool of selected donors and administered via an enema system. The most advanced is a lyophilized powder that can be reconstituted by rectal infusions developed by CIPAC Therapeutics.

4.3 Precision microbiome reconstitution

The lack of regulatory protocol and stiff resistance from clinicians treating chronically ill patients has dampened efforts to introduce FMT as a viable therapy. This led to the development of the concept of “precision microbiome reconstitution,” where a single bacterium can be used to restore colonization resistance in C. difficile patients [105]—providing a more targeted approach where a consortia of specific bacterial strains are identified to treat a particular diseased condition, and this will enable greater specificity and quality control. In germ-free mice, a murine isolate belonging to the family Lachnospiraceae partially restored colonization resistance against C. difficile [165]. An elaborate study using mouse models, clinical studies, metagenomic analysis, and mathematical modeling identified C. scindens as an intestinal bacterium associated with resistance to C. difficile. C. scindens produces growth inhibitory or spore germination inhibitory secondary bile acids to inhibit C. difficile. Furthermore, colonic induction of anti-inflammatory T regulatory cells can be used to develop immunity against dysbiotic conditions. A community of 17 strains including C. scindens induced the development of anti-inflammatory T regulatory cells, and this reduced colitis [67]. They also identified that the concentration of short-chain fatty acids increased upon the colonization of these 17 isolates. The fact that short-chain fatty acids modulated a Treg cell response suggested a common pathway by which different microbes modulated an induction of Treg cells. This opportunity was utilized to identify many more strains mostly belonging to bacteroides that are capable to induce an immune response that can restore colonization resistance from a dysbiotic condition [68]. However, even though a single strain may be able to resist a single organism of interest, a community of organisms reflecting the diversity of microbiota might be needed to restore baseline colonization resistance. This specific targeted approach is used by pharmaceutical firms to develop targeted drugs—for example, Seres Therapeutics is developing SER-109—comprising bacterial spores enriched and purified from healthy stool and packaged into capsules. The product can restructure a dysbiotic gut to a healthy microbiome. Vedanta Biosciences are identifying and developing bacterial strains that can suppress chronic gut inflammation. Similarly, a microbial assemblage derived from stool and grown in culture called RePOOPulate has been developed to treat CDI infections.

4.4 The importance of SAFE FMT

Safety of the patient should be of prime importance when an FMT procedure is considered, especially if the patient is having a poor immune status [166, 167]. Middle gut FMT procedures can cause vomiting and aspiration [168]. The nasojejunal tube could put the patient at high risk of aspiration and should be conducted with anesthesia [143]. There is enough evidence that the long-term safety of the patient should be considered as well. Generally, a tested donor fecal sample is used for FMT. However, this carries the disadvantage that unwanted or potentially pathogenic bacterial phenotypes maybe carried from donors to recipients. A particular case was reported where the patient developed new onset obesity after obtaining a stool sample from a heterologous donor [169]. Using the patient’s own stool sample can avoid the problems associated with donor stool samples. Here, a fecal sample of the patient is banked in the hospital before any procedure that requires antibiotic treatment. The banked sample may provide the vital resource to avoid hospital-acquired infections and to replenish the patient’s own microbiota. Preservation of the patient’s own or donor feces pose a second challenge [170]. There are reports that fecal matter from patients with colon cancer promoted tumorigenesis in germ-free and carcinogenic mice. Potential cardiometabolic, autoimmune, and neurological disease also have been discussed. All these points to the tough screening and regulations are needed before a donor is selected for fecal sample prior FMT.

However, recent reports suggest that FMT is gaining wide acceptance among patients. A survey showed that among patients of Crohn’s disease who received FMT, 56% showed satisfactory clinical efficacy, 74% showed willingness for a second FMT, and 89% expressed willingness to recommend FMT to other patients [171]. Also, the cost efficacy of FMT has been demonstrated worldwide [172, 173, 174, 175, 176, 177]. FMT is still very far from being implemented into routine therapy. The technique needs to undergo rigorous process of standardization before the therapy becomes applied in daily practice. Nevertheless the importance of gut microbiota in maintenance of a healthy lifestyle is demonstrated without doubt. In future therapeutic approaches including antibiotic therapy should take into consideration the impact it has on the gut microbiota and the clinicians should be mindful of the impact of the devastating secondary effects of these therapeutic approaches on the patient.

References

  1. 1. Human Microbiome Project C. A framework for human microbiome research. Nature. 2012;486(7402):215-221
  2. 2. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59-65
  3. 3. Halwachs B, Madhusudhan N, Krause R, Nilsson RH, Moissl-Eichinger C, Hogenauer C, et al. Critical issues in mycobiota analysis. Frontiers in Microbiology. 2017;8:180
  4. 4. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337-340
  5. 5. Neish AS. Microbes in gastrointestinal health and disease. Gastroenterology. 2009;136(1):65-80
  6. 6. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207-214
  7. 7. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nature Reviews. Microbiology. 2016;14(1):20-32
  8. 8. Duncan SH, Louis P, Flint HJ. Cultivable bacterial diversity from the human colon. Letters in Applied Microbiology. 2007;44(4):343-350
  9. 9. O'Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Reports. 2006;7(7):688-693
  10. 10. Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3(4):289-306
  11. 11. LeBlanc JG, Milani C, de Giori GS, Sesma F, van Sinderen D, Ventura M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Current Opinion in Biotechnology. 2013;24(2):160-168
  12. 12. Duncan SH, Scott KP, Ramsay AG, Harmsen HJ, Welling GW, Stewart CS, et al. Effects of alternative dietary substrates on competition between human colonic bacteria in an anaerobic fermentor system. Applied and Environmental Microbiology. 2003;69(2):1136-1142
  13. 13. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nature Reviews. Immunology. 2009;9(5):313-323
  14. 14. Sommer F, Backhed F. The gut microbiota—Masters of host development and physiology. Nature Reviews. Microbiology. 2013;11(4):227-238
  15. 15. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host & Microbe. 2015;17(5):662-671
  16. 16. Reinhardt C, Bergentall M, Greiner TU, Schaffner F, Ostergren-Lunden G, Petersen LC, et al. Tissue factor and PAR1 promote microbiota-induced intestinal vascular remodelling. Nature. 2012;483(7391):627-631
  17. 17. Luna RA, Foster JA. Gut brain axis: Diet microbiota interactions and implications for modulation of anxiety and depression. Current Opinion in Biotechnology. 2015;32:35-41
  18. 18. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. The Journal of Clinical Investigation. 2015;125(3):926-938
  19. 19. Mu C, Yang Y, Zhu W. Gut microbiota: The brain peacekeeper. Frontiers in Microbiology. 2016;7:345
  20. 20. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nature Reviews. Microbiology. 2012;10(11):735-742
  21. 21. Carmody RN, Gerber GK, Luevano JM Jr, Gatti DM, Somes L, Svenson KL, et al. Diet dominates host genotype in shaping the murine gut microbiota. Cell Host & Microbe. 2015;17(1):72-84
  22. 22. Zarrinpar A, Chaix A, Yooseph S, Panda S. Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Cell Metabolism. 2014;20(6):1006-1017
  23. 23. Thaiss CA, Zeevi D, Levy M, Zilberman-Schapira G, Suez J, Tengeler AC, et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell. 2014;159(3):514-529
  24. 24. DeGruttola AK, Low D, Mizoguchi A, Mizoguchi E. Current understanding of dysbiosis in disease in human and animal models. Inflammatory Bowel Diseases. 2016;22(5):1137-1150
  25. 25. Hyman RW, Fukushima M, Diamond L, Kumm J, Giudice LC, Davis RW. Microbes on the human vaginal epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(22):7952-7957
  26. 26. Zhou X, Brown CJ, Abdo Z, Davis CC, Hansmann MA, Joyce P, et al. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. The ISME Journal. 2007;1(2):121-133
  27. 27. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(Suppl 1):4680-4687
  28. 28. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(26):11971-11975
  29. 29. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009;326(5960):1694-1697
  30. 30. Fanaro S, Chierici R, Guerrini P, Vigi V. Intestinal microflora in early infancy: Composition and development. Acta Paediatrica. Supplement. 2003;91(441):48-55
  31. 31. Favier CF, Vaughan EE, De Vos WM, Akkermans AD. Molecular monitoring of succession of bacterial communities in human neonates. Applied and Environmental Microbiology. 2002;68(1):219-226
  32. 32. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS Biology. 2007;5(7):e177
  33. 33. Mackie RI, Sghir A, Gaskins HR. Developmental microbial ecology of the neonatal gastrointestinal tract. The American Journal of Clinical Nutrition. 1999;69(5):1035S-1045S
  34. 34. Coppa GV, Zampini L, Galeazzi T, Gabrielli O. Prebiotics in human milk: A review. Digestive and Liver Disease. 2006;38(Suppl 2):S291-S294
  35. 35. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480-484
  36. 36. Biasucci G, Benenati B, Morelli L, Bessi E, Boehm G. Cesarean delivery may affect the early biodiversity of intestinal bacteria. The Journal of Nutrition. 2008;138(9):1796S-1800S
  37. 37. Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C. Mode of delivery affects the bacterial community in the newborn gut. Early Human Development. 2010;86(Suppl 1):13-15
  38. 38. Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B, Stelma F, et al. Gut microbiota composition and development of atopic manifestations in infancy: The KOALA Birth Cohort Study. Gut. 2007;56(5):661-667
  39. 39. Bager P, Wohlfahrt J, Westergaard T. Caesarean delivery and risk of atopy and allergic disease: Meta-analyses. Clinical and Experimental Allergy. 2008;38(4):634-642
  40. 40. Albenberg L, Esipova TV, Judge CP, Bittinger K, Chen J, Laughlin A, et al. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology. 2014;147(5):1055-1063 e1058
  41. 41. Johansson ME, Larsson JM, Hansson GC. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(Suppl 1):4659-4665
  42. 42. Tropini C, Earle KA, Huang KC, Sonnenburg JL. The gut microbiome: Connecting spatial organization to function. Cell Host & Microbe. 2017;21(4):433-442
  43. 43. Berry D, Stecher B, Schintlmeister A, Reichert J, Brugiroux S, Wild B, et al. Host-compound foraging by intestinal microbiota revealed by single-cell stable isotope probing. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(12):4720-4725
  44. 44. Pedron T, Mulet C, Dauga C, Frangeul L, Chervaux C, Grompone G, et al. A crypt-specific core microbiota resides in the mouse colon. MBio. 2012;3(3):e00116-12
  45. 45. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al. Succession of microbial consortia in the developing infant gut microbiome. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(Suppl 1):4578-4585
  46. 46. Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host & Microbe. 2015;17(5):690-703
  47. 47. Nieuwdorp M, Gilijamse PW, Pai N, Kaplan LM. Role of the microbiome in energy regulation and metabolism. Gastroenterology. 2014;146(6):1525-1533
  48. 48. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031
  49. 49. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022-1023
  50. 50. Sonnenburg JL, Backhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535(7610):56-64
  51. 51. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122(1):107-118
  52. 52. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446-450
  53. 53. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569-573
  54. 54. Stecher B, Maier L, Hardt WD. 'Blooming' in the gut: How dysbiosis might contribute to pathogen evolution. Nature Reviews. Microbiology. 2013;11(4):277-284
  55. 55. Mushin R, Dubos R. Colonization of the mouse intestine with Escherichia coli. The Journal of Experimental Medicine. 1965;122(4):745-757
  56. 56. Bohnhoff M, Drake BL, Miller CP. Effect of streptomycin on susceptibility of intestinal tract to experimental Salmonella infection. Proceedings of the Society for Experimental Biology and Medicine. 1954;86(1):132-137
  57. 57. Bohnhoff M, Miller CP. Enhanced susceptibility to Salmonella infection in streptomycin-treated mice. The Journal of Infectious Diseases. 1962;111:117-127
  58. 58. Freter R. The fatal enteric cholera infection in the guinea pig, achieved by inhibition of normal enteric flora. The Journal of Infectious Diseases. 1955;97(1):57-65
  59. 59. Hentges DJ, Freter R. In vivo and in vitro antagonism of intestinal bacteria against Shigella flexneri. I. Correlation between various tests. The Journal of Infectious Diseases. 1962;110:30-37
  60. 60. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(13):5354-5359
  61. 61. Kommineni S, Bretl DJ, Lam V, Chakraborty R, Hayward M, Simpson P, et al. Bacteriocin production augments niche competition by enterococci in the mammalian gastrointestinal tract. Nature. 2015;526(7575):719-722
  62. 62. Milshteyn A, Schneider JS, Brady SF. Mining the metabiome: Identifying novel natural products from microbial communities. Chemistry & Biology. 2014;21(9):1211-1223
  63. 63. Petersson J, Schreiber O, Hansson GC, Gendler SJ, Velcich A, Lundberg JO, et al. Importance and regulation of the colonic mucus barrier in a mouse model of colitis. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2011;300(2):G327-G333
  64. 64. Earle KA, Billings G, Sigal M, Lichtman JS, Hansson GC, Elias JE, et al. Quantitative imaging of gut microbiota spatial organization. Cell Host & Microbe. 2015;18(4):478-488
  65. 65. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461(7268):1282-1286
  66. 66. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485-498
  67. 67. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232-236
  68. 68. Faith JJ, Ahern PP, Ridaura VK, Cheng J, Gordon JI. Identifying gut microbe-host phenotype relationships using combinatorial communities in gnotobiotic mice. Science Translational Medicine. 2014;6(220):220ra211
  69. 69. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, Koren O, et al. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science. 2011;334(6053):255-258
  70. 70. Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity. 2012;37(1):158-170
  71. 71. Nardone G, Compare D, Rocco A. A microbiota-centric view of diseases of the upper gastrointestinal tract. Lancet Gastroenterology & Hepatology. 2017;2(4):298-312
  72. 72. Hunt RH, Yaghoobi M. The esophageal and gastric microbiome in health and disease. Gastroenterology Clinics of North America. 2017;46(1):121-141
  73. 73. Di Pilato V, Freschi G, Ringressi MN, Pallecchi L, Rossolini GM, Bechi P. The esophageal microbiota in health and disease. Annals of the New York Academy of Sciences. 2016;1381(1):21-33
  74. 74. Yang L, Lu X, Nossa CW, Francois F, Peek RM, Pei Z. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology. 2009;137(2):588-597
  75. 75. Liu N, Ando T, Ishiguro K, Maeda O, Watanabe O, Funasaka K, et al. Characterization of bacterial biota in the distal esophagus of Japanese patients with reflux esophagitis and Barrett's esophagus. BMC Infectious Diseases. 2013;13:130
  76. 76. Blackett KL, Siddhi SS, Cleary S, Steed H, Miller MH, Macfarlane S, et al. Oesophageal bacterial biofilm changes in gastro-oesophageal reflux disease, Barrett's and oesophageal carcinoma: Association or causality? Alimentary Pharmacology & Therapeutics. 2013;37(11):1084-1092
  77. 77. Macfarlane S, Furrie E, Macfarlane GT, Dillon JF. Microbial colonization of the upper gastrointestinal tract in patients with Barrett's esophagus. Clinical Infectious Diseases. 2007;45(1):29-38
  78. 78. Benitez AJ, Hoffmann C, Muir AB, Dods KK, Spergel JM, Bushman FD, et al. Inflammation-associated microbiota in pediatric eosinophilic esophagitis. Microbiome. 2015;3:23
  79. 79. Falush D, Wirth T, Linz B, Pritchard JK, Stephens M, Kidd M, et al. Traces of human migrations in Helicobacter pylori populations. Science. 2003;299(5612):1582-1585
  80. 80. Arnold IC, Dehzad N, Reuter S, Martin H, Becher B, Taube C, et al. Helicobacter pylori infection prevents allergic asthma in mouse models through the induction of regulatory T cells. The Journal of Clinical Investigation. 2011;121(8):3088-3093
  81. 81. Wang M, Ahrne S, Jeppsson B, Molin G. Comparison of bacterial diversity along the human intestinal tract by direct cloning and sequencing of 16S rRNA genes. FEMS Microbiology Ecology. 2005;54(2):219-231
  82. 82. Hartman AL, Lough DM, Barupal DK, Fiehn O, Fishbein T, Zasloff M, et al. Human gut microbiome adopts an alternative state following small bowel transplantation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(40):17187-17192
  83. 83. Booijink CC, El-Aidy S, Rajilic-Stojanovic M, Heilig HG, Troost FJ, Smidt H, et al. High temporal and inter-individual variation detected in the human ileal microbiota. Environmental Microbiology. 2010;12(12):3213-3227
  84. 84. Caminero A, Galipeau HJ, McCarville JL, Johnston CW, Bernier SP, Russell AK, et al. Duodenal bacteria from patients with celiac disease and healthy subjects distinctly affect gluten breakdown and immunogenicity. Gastroenterology. 2016;151(4):670-683
  85. 85. Staffas A, Burgos da Silva M, van den Brink MR. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood. 2017;129(8):927-933
  86. 86. van Bekkum DW, Roodenburg J, Heidt PJ, van der Waaij D. Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora. Journal of the National Cancer Institute. 1974;52(2):401-404
  87. 87. Jones JM, Wilson R, Bealmear PM. Mortality and gross pathology of secondary disease in germfree mouse radiation chimeras. Radiation Research. 1971;45(3):577-588
  88. 88. Taur Y, Jenq RR, Perales MA, Littmann ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124(7):1174-1182
  89. 89. Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, Peter K, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: Loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biology of Blood and Marrow Transplantation. 2014;20(5):640-645
  90. 90. Dethlefsen L, McFall-Ngai M, Relman DA. An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature. 2007;449(7164):811-818
  91. 91. Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M, et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. The Journal of Clinical Investigation. 2010;120(12):4332-4341
  92. 92. Buffie CG, Jarchum I, Equinda M, Lipuma L, Gobourne A, Viale A, et al. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infection and Immunity. 2012;80(1):62-73
  93. 93. Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nature Reviews. Immunology. 2013;13(11):790-801
  94. 94. Vollaard EJ, Clasener HA, Janssen AJ. Decontamination of the bowel by intravenous administration of pefloxacin. The Journal of Antimicrobial Chemotherapy. 1990;26(6):847-852
  95. 95. Vollaard EJ, Clasener HA, van Griethuysen AJ, Janssen AJ, Sanders-Reijmers AJ. Influence of amoxycillin, erythromycin and roxithromycin on colonization resistance and on appearance of secondary colonization in healthy volunteers. The Journal of Antimicrobial Chemotherapy. 1987;20(Suppl B):131-138
  96. 96. Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. The ISME Journal. 2007;1(1):56-66
  97. 97. Dominguez-Bello MG, Blaser MJ, Ley RE, Knight R. Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology. 2011;140(6):1713-1719
  98. 98. Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Reports. 2012;13(5):440-447
  99. 99. Risnes KR, Belanger K, Murk W, Bracken MB. Antibiotic exposure by 6 months and asthma and allergy at 6 years: Findings in a cohort of 1,401 US children. American Journal of Epidemiology. 2011;173(3):310-318
  100. 100. Korpela K, Salonen A, Virta LJ, Kekkonen RA, Forslund K, Bork P, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nature Communications. 2016;7:10410
  101. 101. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158(4):705-721
  102. 102. Britton RA, Young VB. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology. 2014;146(6):1547-1553
  103. 103. van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. The New England Journal of Medicine. 2013;368(5):407-415
  104. 104. Schubert AM, Rogers MA, Ring C, Mogle J, Petrosino JP, Young VB, et al. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. MBio. 2014;5(3):e01021-e01014
  105. 105. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517(7533):205-208
  106. 106. Theriot CM, Koenigsknecht MJ, Carlson PE Jr, Hatton GE, Nelson AM, Li B, et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nature Communications. 2014;5:3114
  107. 107. Hogenauer C, Langner C, Beubler E, Lippe IT, Schicho R, Gorkiewicz G, et al. Klebsiella oxytoca as a causative organism of antibiotic-associated hemorrhagic colitis. The New England Journal of Medicine. 2006;355(23):2418-2426
  108. 108. Schneditz G, Rentner J, Roier S, Pletz J, Herzog KA, Bucker R, et al. Enterotoxicity of a nonribosomal peptide causes antibiotic-associated colitis. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(36):13181-13186
  109. 109. Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annual Review of Immunology. 2010;28:573-621
  110. 110. Orholm M, Munkholm P, Langholz E, Nielsen OH, Sorensen TI, Binder V. Familial occurrence of inflammatory bowel disease. The New England Journal of Medicine. 1991;324(2):84-88
  111. 111. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nature Genetics. 2008;40(8):955-962
  112. 112. Lees CW, Barrett JC, Parkes M, Satsangi J. New IBD genetics: Common pathways with other diseases. Gut. 2011;60(12):1739-1753
  113. 113. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119-124
  114. 114. Becker C, Neurath MF, Wirtz S. The intestinal microbiota in inflammatory bowel disease. ILAR Journal. 2015;56(2):192-204
  115. 115. Castano-Rodriguez N, Kaakoush NO, Lee WS, Mitchell HM. Dual role of Helicobacter and Campylobacter species in IBD: A systematic review and meta-analysis. Gut. 2017;66(2):235-249
  116. 116. Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, et al. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host & Microbe. 2014;15(3):382-392
  117. 117. Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host & Microbe. 2013;14(2):207-215
  118. 118. Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344-355
  119. 119. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host & Microbe. 2013;14(2):195-206
  120. 120. Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66(4):683-691
  121. 121. Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012;338(6103):120-123
  122. 122. Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Research. 2012;22(2):299-306
  123. 123. Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170(3):548-563 e516
  124. 124. Feng Q, Liang S, Jia H, Stadlmayr A, Tang L, Lan Z, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nature Communications. 2015;6:6528
  125. 125. Lawley TD, Clare S, Walker AW, Stares MD, Connor TR, Raisen C, et al. Targeted restoration of the intestinal microbiota with a simple, defined bacteriotherapy resolves relapsing Clostridium difficile disease in mice. PLoS Pathogens. 2012;8(10):e1002995
  126. 126. Andrews PJ, Borody TJ. “Putting back the bugs”: Bacterial treatment relieves chronic constipation and symptoms of irritable bowel syndrome. The Medical Journal of Australia. 1993;159(9):633-634
  127. 127. Tvede M, Rask-Madsen J. Bacteriotherapy for chronic relapsing Clostridium difficile diarrhoea in six patients. Lancet. 1989;1(8648):1156-1160
  128. 128. Zhang F, Luo W, Shi Y, Fan Z, Ji G. Should we standardize the 1,700-year-old fecal microbiota transplantation? American Journal of Gastroenterology. 2012;107(11):1755; author reply p 1755-1756
  129. 129. Ren RR, Sun G, Yang YS, Peng LH, Wang SF, Shi XH, et al. Chinese physicians' perceptions of fecal microbiota transplantation. World Journal of Gastroenterology. 2016;22(19):4757-4765
  130. 130. Eiseman B, Silen W, Bascom GS, Kauvar AJ. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery. 1958;44(5):854-859
  131. 131. Fischer M, Sipe BW, Rogers NA, Cook GK, Robb BW, Vuppalanchi R, et al. Faecal microbiota transplantation plus selected use of vancomycin for severe-complicated Clostridium difficile infection: Description of a protocol with high success rate. Alimentary Pharmacology & Therapeutics. 2015;42(4):470-476
  132. 132. Brandt LJ, Aroniadis OC, Mellow M, Kanatzar A, Kelly C, Park T, et al. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. The American Journal of Gastroenterology. 2012;107(7):1079-1087
  133. 133. Hamilton MJ, Weingarden AR, Sadowsky MJ, Khoruts A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. The American Journal of Gastroenterology. 2012;107(5):761-767
  134. 134. Lee CH, Steiner T, Petrof EO, Smieja M, Roscoe D, Nematallah A, et al. Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent Clostridium difficile infection: A randomized clinical trial. JAMA. 2016;315(2):142-149
  135. 135. Surawicz CM, Brandt LJ, Binion DG, Ananthakrishnan AN, Curry SR, Gilligan PH, et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. The American Journal of Gastroenterology. 2013;108(4):478-498; quiz 499
  136. 136. Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, Daigneault MC, et al. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: 'RePOOPulating' the gut. Microbiome. 2013;1(1):3
  137. 137. Martz SL, Guzman-Rodriguez M, He SM, Noordhof C, Hurlbut DJ, Gloor GB, et al. A human gut ecosystem protects against C. difficile disease by targeting TcdA. Journal of Gastroenterology. 2017;52(4):452-465
  138. 138. Bennet JD, Brinkman M. Treatment of ulcerative colitis by implantation of normal colonic flora. Lancet. 1989;1(8630):164
  139. 139. Kelly CR, Kahn S, Kashyap P, Laine L, Rubin D, Atreja A, et al. Update on fecal microbiota transplantation 2015: Indications, methodologies, mechanisms, and outlook. Gastroenterology. 2015;149(1):223-237
  140. 140. Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet. 2017;389(10075):1218-1228
  141. 141. He Z, Li P, Zhu J, Cui B, Xu L, Xiang J, et al. Multiple fresh fecal microbiota transplants induces and maintains clinical remission in Crohn’s disease complicated with inflammatory mass. Scientific Reports. 2017;7(1):4753
  142. 142. He Z, Cui BT, Zhang T, Li P, Long CY, Ji GZ, et al. Fecal microbiota transplantation cured epilepsy in a case with Crohn's disease: The first report. World Journal of Gastroenterology. 2017;23(19):3565-3568
  143. 143. Cui B, Feng Q, Wang H, Wang M, Peng Z, Li P, et al. Fecal microbiota transplantation through mid-gut for refractory Crohn's disease: Safety, feasibility, and efficacy trial results. Journal of Gastroenterology and Hepatology. 2015;30(1):51-58
  144. 144. Cui B, Li P, Xu L, Peng Z, Xiang J, He Z, et al. Step-up fecal microbiota transplantation (FMT) strategy. Gut Microbes. 2016;7(4):323-328
  145. 145. Cho JA, Chinnapen DJF. Targeting friend and foe: Emerging therapeutics in the age of gut microbiome and disease. Journal of Microbiology. 2018;56(3):183-188
  146. 146. Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931-943
  147. 147. Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97-103
  148. 148. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91-97
  149. 149. Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104-108
  150. 150. Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell. 2018;33(4):570-580
  151. 151. Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079-1084
  152. 152. Ferreira MR, Muls A, Dearnaley DP, Andreyev HJ. Microbiota and radiation-induced bowel toxicity: Lessons from inflammatory bowel disease for the radiation oncologist. The Lancet Oncology. 2014;15(3):e139-e147
  153. 153. Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nature Reviews. Gastroenterology & Hepatology. 2017;14(6):356-365
  154. 154. Cui M, Xiao H, Li Y, Zhou L, Zhao S, Luo D, et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Molecular Medicine. 2017;9(4):448-461
  155. 155. Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clinical Infectious Diseases. 2012;55(7):905-914
  156. 156. Taur Y, Pamer EG. Harnessing microbiota to kill a pathogen: Fixing the microbiota to treat Clostridium difficile infections. Nature Medicine. 2014;20(3):246-247
  157. 157. Ubeda C, Bucci V, Caballero S, Djukovic A, Toussaint NC, Equinda M, et al. Intestinal microbiota containing Barnesiella species cures vancomycin-resistant Enterococcus faecium colonization. Infection and Immunity. 2013;81(3):965-973
  158. 158. Jiang ZD, Ajami NJ, Petrosino JF, Jun G, Hanis CL, Shah M, et al. Randomised clinical trial: Faecal microbiota transplantation for recurrent Clostridum difficile infection—Fresh, or frozen, or lyophilised microbiota from a small pool of healthy donors delivered by colonoscopy. Alimentary Pharmacology & Therapeutics. 2017;45(7):899-908
  159. 159. Chu ND, Smith MB, Perrotta AR, Kassam Z, Alm EJ. Profiling living bacteria informs preparation of fecal microbiota transplantations. PLoS One. 2017;12(1):e0170922
  160. 160. Cammarota G, Ianiro G, Tilg H, Rajilic-Stojanovic M, Kump P, Satokari R, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66(4):569-580
  161. 161. Peng Z, Xiang J, He Z, Zhang T, Xu L, Cui B, et al. Colonic transendoscopic enteral tubing: A novel way of transplanting fecal microbiota. Endoscopy International Open. 2016;4(6):E610-E613
  162. 162. Youngster I, Russell GH, Pindar C, Ziv-Baran T, Sauk J, Hohmann EL. Oral, capsulized, frozen fecal microbiota transplantation for relapsing Clostridium difficile infection. JAMA. 2014;312(17):1772-1778
  163. 163. Zhang F, Amateau SK, Khashab MA, Okolo PI 3rd. Mid-gut stents. Current Opinion in Gastroenterology. 2012;28(5):451-460
  164. 164. Long C, Yu Y, Cui B, Jagessar SAR, Zhang J, Ji G, et al. A novel quick transendoscopic enteral tubing in mid-gut: Technique and training with video. BMC Gastroenterology. 2018;18(1):37
  165. 165. Reeves AE, Koenigsknecht MJ, Bergin IL, Young VB. Suppression of Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae. Infection and Immunity. 2012;80(11):3786-3794
  166. 166. Fischer M, Kao D, Mehta SR, Martin T, Dimitry J, Keshteli AH, et al. Predictors of early failure after fecal microbiota transplantation for the therapy of Clostridium difficile infection: A multicenter study. The American Journal of Gastroenterology. 2016;111(7):1024-1031
  167. 167. Fischer M, Kao D, Kelly C, Kuchipudi A, Jafri SM, Blumenkehl M, et al. Fecal microbiota transplantation is safe and efficacious for recurrent or refractory Clostridium difficile infection in patients with inflammatory bowel disease. Inflammatory Bowel Diseases. 2016;22(10):2402-2409
  168. 168. Gweon TG, Kim J, Lim CH, Park JM, Lee DG, Lee IS, et al. Fecal microbiota transplantation using upper gastrointestinal tract for the treatment of refractory or severe complicated clostridium difficile infection in elderly patients in poor medical condition: The first study in an Asian country. Gastroenterology Research and Practice. 2016;2016:2687605
  169. 169. Alang N, Kelly CR. Weight gain after fecal microbiota transplantation. Open Forum Infectious Diseases. 2015;2(1):ofv004
  170. 170. Hale VL, Tan CL, Niu K, Yang Y, Cui D, Zhao H, et al. Effects of field conditions on fecal microbiota. Journal of Microbiological Methods. 2016;130:180-188
  171. 171. Xu L, Zhang T, Cui B, He Z, Xiang J, Long C, et al. Clinical efficacy maintains patients' positive attitudes toward fecal microbiota transplantation. Medicine (Baltimore). 2016;95(30):e4055
  172. 172. Konijeti GG, Sauk J, Shrime MG, Gupta M, Ananthakrishnan AN. Cost-effectiveness of competing strategies for management of recurrent Clostridium difficile infection: A decision analysis. Clinical Infectious Diseases. 2014;58(11):1507-1514
  173. 173. Varier RU, Biltaji E, Smith KJ, Roberts MS, Kyle Jensen M, LaFleur J, et al. Cost-effectiveness analysis of fecal microbiota transplantation for recurrent Clostridium difficile infection. Infection Control and Hospital Epidemiology. 2015;36(4):438-444
  174. 174. Waye A, Atkins K, Kao D. Cost averted with timely fecal microbiota transplantation in the management of recurrent Clostridium difficile infection in Alberta, Canada. Journal of Clinical Gastroenterology. 2016;50(9):747-753
  175. 175. Merlo G, Graves N, Brain D, Connelly LB. Economic evaluation of fecal microbiota transplantation for the treatment of recurrent Clostridium difficile infection in Australia. Journal of Gastroenterology and Hepatology. 2016;31(12):1927-1932
  176. 176. Baro E, Galperine T, Denies F, Lannoy D, Lenne X, Odou P, et al. Cost-effectiveness analysis of five competing strategies for the management of multiple recurrent community-onset Clostridium difficile infection in France. PLoS One. 2017;12(1):e0170258
  177. 177. Zhang T, Xiang J, Cui B, He Z, Li P, Chen H, et al. Cost-effectiveness analysis of fecal microbiota transplantation for inflammatory bowel disease. Oncotarget. 2017;8(51):88894-88903

Written By

Shabarinath Srikumar and Séamus Fanning

Submitted: 10 August 2018 Reviewed: 13 August 2018 Published: 04 December 2018