5,557

Metagenomic Study of Human Gastrointestinal Tracts in Health and Diseases

Firouz Abbasian, Tayyebeh Saberbaghi

Firouz Abbasian, Tayyebeh Saberbaghi, Department of Microbiology, Science and Research campus, Islamic Azad University, Tehran, Iran

Correspondence to: Firouz Abbasian, Department of Microbiology, Science and Research campus, Islamic Azad University, Tehran, Iran.
Email: Firouz.abbasian@gmail.com
Telephone:+98-911-136-3088
Received: September 16, 2013
Revised: September 28, 2013
Accepted: October 2, 2013
Published online: December 21, 2013

ABSTRACT

Metagenomics is a new emerged technology used to investigate the microbial diversity in an environment and to study their function in a given environment. The Human Microbiome Project is a global project to research diversity of microbial communities associated with human body and to investigate their effects on physiology of organs. These studies also are interested to link between the existence of a microbial population and several inflammatory, physiological, metabolic and psychological disorders. Based on metagenomic studies, this paper aims to review the newest information regarding microbial diversity of the intestinal tracts and their physiologic and/or pathologic influences on human body.

Key words: Metagenomics; Biodiversity; Intestinal flora; Malnutrition; Physiological disorders; Psychological disorders

© 2013 The Authors. Published by ACT Publishing Group Ltd.

Abbasian F, Saberbaghi T. Metagenomic Study of Human Gastrointestinal Tracts in Health and Diseases. Journal of Gastroenterology and Hepatology Research 2013; 2(12): 885-896 Available from: URL: http://www.ghrnet.org/index.php/joghr/article/view/546

Introduction

Employment of new emerged detecting technologies, referred to as metagenomics, has changed our view of microbial diversity in different habitats, including the micro-flora of gastrointestinal tracts. Metagenomics is an approach in which whole genome contents of a community of (micro) organisms in a niche of interest is investigated in order to detect its microbial diversity and also to study special trait(s) of the habitat[1,2]. This technique employs the hyper-variable sections of special marker genes, especially the 16S ribosomal RNA (rRNA), to identify microbial diversity. Also, this technique is able to detect the functional ability of existing microorganisms in an environment based on the identified genes[3]. The metagenomic approach can be used for any natural environment where microbial genomic sources are available. The Human Microbiome Project is an international study of the microbial communities associated with different parts of our body[4]. Most of the existing information regarding the microbial diversity of human body and their role in different physiological functions of the gastrointestinal tract are based on in vitro or culture based studies. However, the new approaches have opened new windows for understanding these relationships between human cells and microbial strains.

The gastrointestinal tract consists of different parts, including mouth, esophagus, stomach, small intestine, colon and rectum. These tracts function as digestive and absorptive organ for our body and at the same time are known as a major exocrine and endocrine hormone producer and an important immune organs[5,6]. Based on the conventional culture-based approaches, a few hundred microorganisms has been detected at different parts of gastrointestinal tracts and the population of obligate anaerobic bacteria, specially Bacteroidetes, are one thousands time more than the population of facultative organisms. However, based on culture-independent approaches 1,012 cells lives in 1g stool, consisting of over 13,000 microbial species that most of them belonged to the Firmicutes and Bacteroidetes (respectively, 75% and 16%)[7,8]. This number in some papers is suggested more than 1,800 genera and 15,000 to 36,000 species, varying based on the bacterial classification system[9]. Only 20% of these microorganisms are detectable by conventional culture-based methods[7]. Although the diversity may differ in each individual person or society, the ecological niche of the microbial combination is similar[10]. In this paper, diversity and function of normal flora of the gastrointestinal tracts is reviewed based on the newest information obtained from culture independent technologies.

THE NORMAL FLORA OF INTESTINAL TRACT AT BIRTH

While intestinal tract is sterile at the birth, it may be contaminated through birth[11]. Although based on researches in 1990s, mode of delivery (normal vaginal birth or caesarean section) had no significant effect(s) on the normal flora of the neonatal intestinal tracts[12,13], nowadays researchers believe in an entirely different way. In normal vaginal delivery, the neonate is affected by both mother’s vaginal and intestinal flora, and the neonate’s gut is colonized predominantly by E. coli, Enterococci, Bifidobacterium, Lactobacillus and Prevotella[14-16]. However, since the neonates delivered via caesarean mode are not exposed to the mother’s vaginal/intestinal flora, their intestinal microbial composition is similar to the microbial flora of skin, consisting of a lower microbial diversity in which Staphylococcus is a dominant population[14,15].

In a culture based research, it has been shown that the mode of feeding has fundamental effects on the normal flora of the intestine at the first days of birth[17]. Approximately 24 hours after a normal birth, the intestinal tract acquire some microorganisms originated from mother’s vagina or her intestinal tracts through delivery or taking milk[18]. Regardless of mode of feeding, the intestine is occupied mostly by E. coli, also some other enterobacteriaceae and gram-positive cocci by the end of first week[12]. Microarray based investigations showed that the microbial population of intestine is formed after one week, but the flora will not reach to its equilibrium by the end of infancy[18]. These flora constitutes mainly of Bifidobacterium sp., E. coli, Enterococcus sp., Streptococcus sp., Staphylococcus sp., Actinomyces sp., Clostridium sp. and Bacteroides sp[19, 20]. It has been shown that roughly 6 day after birth the intestinal tracts of breastfed and bottle-fed infants were dominantly colonized by Bifidobacteria and enterobacteriaceae, respectively. These conditions continued by the end of one month when Bifidobacteria were the dominant bacteria in both groups. In this age, however, the number of Bifidobacteria in the stool of bottle-fed infants was 1/10 in comparison to breast-fed infants[17]. In a study performed based on 16S rRNA amplification, Bifidobacterium sp. was dominant member of the fecal samples taken from infants below four month old, and the number of this bacteria raised from 45% in eight days old infants to 64% in 117 days old infants[21]. It is important to be mentioned that since nutrition shortage affects directly mother’s normal flora and her immune system[22], it is suggested that malnutrition before and after delivery might change indirectly the neonate’s microbiota. Furthermore, this condition affects milk components in terms of nutrients and different immunoactive compounds such as antimicrobial enzymes, Interleukin 10 (IL10) and several growth factors[22].

THE ORAL CAVITY

The oral cavity provides different special habits where vast variety of microorganisms are colonized[23]. Based on information in the Human Oral Microbiome Database (http://www.homd.org/) approximately 688 species of prokaryotes, including only one species of archaea (Methanobrevibacter oralis), have been characterized that only 49% of them are known microorganisms. These bacteria belong to different phyla, including Firmicutes (33.9%), Proteobacteria (16.57%), Bacteroidetes (17.73%), Actinobacteria (13.23%), Spirochaetes (7.12%), Fusobacteria (5.38%), TM7 (1.75%), Tenericutes (1.6)%, Synergistetes (1.45%), SR1 (0.44%), GN02 (0.44%), Chloroflexi (0.15%) and Chlamydiae (0.15%) (Table 1)[24]. The composition and abundance of microorganisms in different part of oral cavity is varied. For instance, hard palate, keratinized gingiva and buccal mucosa are occupied with Firmicutes (mostly Streptococcus sp. and Gemella sp.) followed by Proteobacteria, Bacteroidetes, Actinobacteria and Fusobacteria in a diminishing order[25]. Also, throat, tonsils, tongue and saliva are inhabited mostly by Firmicutes, especially Streptococcus sp., Veillonella sp. and Lachnospiraceae (Oribacterium sp. and Catonella sp.) followed by Bacteroidetes (Prevotella sp.), Neisseria sp., Fusobacteria (Fusobacterium sp. and Leptotrichia sp.), Actinobacteria (Actinomyces sp.) and TM7 in a diminishing order. However, the abundance of Firmicutes in the plaques formed on both supra-and sub-gingival habitats is decreased but the number of Actinobacteria shows a significant increase[25]. In addition to the strains found in the throat and tongue, these last habitat are occupied by Rothia sp., Corynebacterium sp., Porphyromonas sp. and Capnocytophaga sp[25]. The pathogenic microorganisms involved in oral and systemic infections like Treponema sp., Aggregatibacter sp., Megasphaera sp., S. pyogenes, S. pneumoniae, H. influenzae and N. meningitidis are also found in those area in low abundance[25].

ESOPHAGUS

The esophagus is a unique part of gastrointestinal tracts since foods are not deposing in the tract. Based on the culture dependent methods using the samples given from luminal washes of the tract, most researches showed a sterile environment where sometimes is contaminated by the transit microorganisms originating from upstairs or downstairs organs[26] like Herpesvirus, Cryptococcus sp. and Candida sp[28-31]. However, based on 16SrRNA pyrosequencing, esophagus is occupied by a stable flora consisting of same series of microorganisms as the oral cavity and nasal swab and roughly 140 species from six phylum of the microorganisms Firmicutes (mostly Streptococcus sp. and Veillonellance), Bacteroides (mostly Prevotella sp.), Actinobacteria, Proteobacteria, Fusobacteria, and TM7[27-29]. The habitat is dominantly occupied by Streptococci (78% of the microbial population)[27]. The composition of esophageal flora change in different physiologic and pathologic conditions, such as gastro-esophageal reflux disease (GERD) and Barrett’s esophagus. In these pathologic and inflammatory conditions, the tract is more hospitable for anaerobic and microaerophilic bacteria. For instance, it has been shown that the staphylococci population is declined to 29% in Barrett’s esophagus[27]. Also, 16sRNA assay showed that the normal flora of this area is changed from 10 to 5 species in the Barrett’s esophagus and 17 species in the reflux esophagitis patients[30].

THE STOMACH

The acid secretion into the stomach makes the tract as special micro-ecosystems, including the gastric mucosa-associated and the gastric juice-associated habitats, where many bacteria cannot survive. Although many microorganisms have been found in the habitats, many of them are transient and came by swallowed foods. Based on 16SrRNA sequencing, however, several acid resistant microorganisms like Helicobacter sp., Prevotella sp., Lactobacillus sp., Streptococcus sp., Enterococcus sp. Staphylococcus sp. Stomatococcus sp., Pseudomonas sp., Rothia sp. and Neisseria sp., originated from both upper and lower intestinal tracts, are normally isolated from these areas[31]. The number of microorganisms inhabited in the stomach is increasing by using new metagenomic approaches, and up to now it is claimed that 13 classes of microorganisms, from 262 phylotypes, live in the stomach. Despite a relatively stable flora inhabited in the gastric mucosa-associated, micro-flora of gastric juice-associated environment is affected by the swallowed foods[32].

It is believed that the non-H. pylori microorganisms in the gastric environments function as antigenic stimulators and increase the abnormality caused by H. pylori[33]. Furthermore, based on animal studies, it has been suggested that intragastric flora assist H. pylori to cause gastric cancer as a result of increase in repairing rate of mucosal injury, and therefore, due to increase in cellular replication[34,35]. Also, activity of some microorganisms in the area, such as Eubacterium limosum, enhance colonization of H. pylori in the area[36,37]. However, the relationship between H. pylori and other gastric microorganisms is not always friendly. For instance, several studies have shown that probiotic microorganisms, such as Bifidobacterium sp., Lactobacillus sp. and Saccharomyces sp. are able to decline the inflammation caused by H. pylori through prevention of microbial colonization, eradication of H. pylori, decrease the side effects caused by administration of antibiotics and finally by reduction of relapse rates[38-40]. On the other hand, stabilization of H. pylori in the gastric environments amends distribution and number of lactobacilli[41].

INTESTINAL TRACTS

The small intestine consists histologically of three different parts, namely duodenum, jejunum and ileum. Due to inhibitory effects caused by acidic pH (of gastric juices), bile salts (released from gallbladder), IgA (immunoglobulin A), the antimicrobial compounds (released from small intestine like C-type lectins, cathelicidins and defensins), little amounts of microbes (less than 103) have been isolated from duodenum[42-44]. In addition, several physiological conditions, such as peristalsis movement of the tract, desquamation of epithelial cells and mucosal flow speed up the transient period of foods through the trace (between 30 minute to 3.5 hours) and thus wash the microbial cells up from this tracts[45]. The number of microorganisms raises through the small intestine and reach to 108 microorganisms per milliliter intestinal juice due to decrease in the level of the inhibitory factors[46]. For instance, alkaline secretions from pancreas lead to decrease of pH profile in a healthy person from 5.5-7.0 in the duodenum to 6.5-7.5 in the ileum[47]. Furthermore, metagenomic studies showed that the bile salt hydrolysis (BSH) activities enhance bacterial resistance to the Conjugated bile acids (CBAs), facilitating their colonization in the gut[48].

Based on the NIH human microbiome project (HMP), the microbiota in oral cavity is more diverse than small and large intestines. In the other word, no members of Chlamydiae, Chloroflexi, GN02, Spirochaetes, SR1, Tenericutes and TM7 as well as the members of Alpha proteobacteria have been found in the healthy human gut (Table 2) (http://www.hmpdacc.org/catalog/). In upper parts of the small intestine, most habitats are occupied by acid tolerant streptococci and lactobacilli[49]. Due to the physical, chemical and physiological changes through the small intestine, the dominant members of microbial community in jejunum shifts to class bacilli, mostly Streptococci, and Proteobacteria[46]. Normal flora of the ileum is similar to colon and contains more than 1,013 microbes, consisting of 500 different species[50]. While the Enterobacteriaceae are dominant microorganisms within the lumen of small intestine, the majority of microorganisms identified in this intestinal mucosa of this area consist of the Clostridiales clusters XIV, XI and IV and Bacteroidetes[51-53].

Due to low concentration of the inhibitory factors, lack of Peyer’s patches and lower peristalsis of the large intestinal tract (and longer retention time of food in the tract), the large intestine (colon) contains higher microbial population than small intestine[54]. The colon is populated with a big community (1×1013-1×1014 cells per gram stool) of microorganisms (approximately 10-30 percentage of stool weight), consisting of more than 500 species and around 2×106 genes (100 times more than the human genome)[55,56]. Normal flora of stool consists frequently of Bacteroidetes (especially Bacteroides sp.), Firmicutes, Proteobacteria, Actinobacteria and Fusobacteria in decreasing order[25]. However, 30-40 species of microbial population in stools constitute 99% of the population. Although the habitat is occupied mainly by Firmicutes (clusters IX, XIV, and XVI) and Bacteroidetes (35%) (especially, Bacteroides, Prevotella and Faecalibacterium), other bacterial phyla, including Proteobacteria (13-15%) and Fusobacteria (7-8%), Actinobacteria, Verrucomicrobia as well as few archaea have been also found in this tract[7,57,58].

Furthermore, the habitat is occupied by approximately 50 fungal phylogroups (like Saccharomyces sp., Galactomyces sp., Penicillium sp., Candida sp., Gloeotinia sp. and Paecilomyces sp.)[59,60]. Based on 18S rRNA studies, Blastocystis sp subtypes II, III and IV are dominant fungi in the distal intestinal tract[60]. While few numbers of protozoans have been adapted themselves to live in host associated niches, these microorganisms can be found in a vast variety of vertebrate hosts. By now, several types of parasite or commensal protozoa like Endolimax sp., Entamoeba histolytica, Entamoeba coli, Entamoeba invadens, Iodamoeba sp., Trichomonas hominis, Chilomastix sp., Pentatrichomonas sp. and Giardia intestinalis have been isolated in human gut[61,62]. Although these protozoa are distributed worldwide, their prevalence in a geographic area is mostly dependent to poor sanitation of inhabitants[63]. Also, while existence of Cyanobacteria in human gut has been proven by molecular studies, no cyanobacterial strain has been isolated yet[64]. Methanobrevibacter smithii and Methanosphaera stadtmanae are the only confirmed archaea isolated from human gut. However, some other archaea like Methanobrevibacter oralis (the only isolated archaea from saliva), Methanosarcina, Stadtmanae and Methanosphaera have been occasionally isolated from these area[65]. Also, presence of low abundant of some pathogenic microorganisms, such as H. pylori in the lower intestinal tracts has been proven[25]. Despite the homology between bacterial population of the intestine, intestinal virome is unique to each person and 95% of this viral diversity is retained in individuals over times[61].

Based on the experiments conducted with new emerged techniques, the intestinal microbiome participate in several activities, including normal cell differentiation and development of the intestinal tracts, tissue homeostasis, natural defence system against pathogenic microorganisms of the tract, host energy metabolism and as an effective facilitating factor for metabolism of different swallowed foods, complex hydrocarbons and fibres[6,66,67]. These flora assist the body to keep its health as a result of production of short chain alcohols and acids, production of vitamin K and B family, alteration in bile salt composition, degradation of food and preparation of a better condition for adsorption of nutrients by intestinal cells[11,66]. Although each society may show a special microbial diversity profile in their intestinal tracts, there is a core gut microbiome in all investigated societies which totally harbour a similar collection of genes. Indeed, as far as a diversity of microorganisms is able to provide the special niche required in a healthy intestinal tracts, it can efficiently work for the body. However, the slight alterations in the phylotype composition of a person, due to excitement, hunger, nutrition, antibiotic treatment and infection, leads to an unique ecological condition that may intensify susceptibility of a person to infectious or physiological disorders[10,20,57].

By now, several physiological features like energy metabolism[11,68,69], degradation of xenobiotics and drugs[70,71], cell differentiation and development[6,72], maturation and activity of immune system[73,74] and the host response to damages in the intestinal epithelial cells have been attributed to the gut microbiome[75,76]. For instance, the intestinal tracts of germ-free mice showed abnormal long intestinal villi[6,72], abnormal enlarged cecum and altered gastrointestinal motion[6]. In this hypothesis, the SCFAs (short chain fatty acids) produced by commensal bacteria play as an immune system modular through two different mechanism: activation of cellular GPRs (G-coupled-protein-receptor) and inactivation of histone deacetylase (HDAC), thus leading to the immunological responses seen in autoimmune disorders such as atopy and asthma[77,78]. Based on studies on germ free rats, the commensal bacteria are essential for development of gastrointestinal-associated lymphoid tissue (GALT) and IECs (intestinal epithelial cells) in the small intestine[73,74]. The IECs use TLRs (Toll Like Receptors), PRPs (Pattern Recognition Receptors) and NLRs (Nod Like Receptors) to differentiate pathogens from commensal microorganism[76]. Since the constitution of microbial society in the intestinal tracts of preterm neonates who are kept in incubator is delayed in compare to the healthy neonates, the intestinal tract of these infants show slow peristalsis and the neonates are severely susceptible to different intestinal infectious disorders[79]. Also, it has been shown that the intestine of germ free mice do not produce angiogenin 4, delaying the formation of villus capillaries in their small intestine[68,80]. Furthermore, it has been shown that the intestinal flora are able to induce production of the transcription factor NF-κB[81]. The NF-κB released into the intestinal tracts induces expression of a variety of genes involved in cell proliferation, cell differentiation and pro-inflammatory responses through infection. Overexpression or deficiency of these factors may lead to occurrence of several pathogenic conditions like obesity and chronic IBD (inflammatory bowel disease)[82-84].

Since hosts live with this flora for millions of years and different types of ecological relationship have been established between host and the microorganisms, it is suggested that any factor that affects these relationships might lead to pathologic conditions. Overall, the balance in population of microbial flora is critical for our health and according to hygiene hypothesis, it is believed that imbalanced microbial population in our body due to improvement in public health is a potential etiological factor for several intestinal pathogenic situations like inflammatory bowel diseases (IBDs), circulatory disease, obesity and autism to the microbiota, atopy and asthma[85-92]. Studies on several intestinal associated diseases or even many systemic disorders have indicated intensive changes in microbial diversity and their composition. For instance, studies on CD (Coeliac disease) showed that the microbial diversity, especially in the proportion of members of Firmicutes decreased significantly while there were highly increases in the number of bacterial population, particularly in Gram negative bacteria. At the same time, the ratio of Bacteroides-E. coli (shigella) to Lactobacillus-Bifidobacterium was higher in these patients[90-92]. It has been shown that overgrowth of E. coli in the gut lead to LPS-TLR4 (Toll like receptor 4) signalling in the gut and therefore to trigger an inflammatory response[93]. Antibiotic associated disorders are well-known clinical problems in which oral administration of some broad-spectrum antibiotic, such as clindamycin, ampicillin, neomycin, erythromycin, metronidazole and cephalosporins alter temporarily the aerobic flora of intestinal tract mostly to Clostridiales order like Subdoligranulum, Acetivibrio, Butyricicoccus, Dorea, Collinsella, leading to severe intestinal diseases such as antibiotic-associated diarrhea and pseudomembranous colitis[94].

Although it has not been clearly proven, intestinal microorganisms can function as a potent etiology of colorectal cancer (CRC) due to release of free radicals and genotoxins and also as a result of induction of Th dependent cell proliferation and TLR dependent procarcinogenic mechanisms[95]. Based on 16S rDNA DGGE analysis, a significant increase in diversity of microorganisms, especially in Clostridium coccoides and Clostridium leptum, was recorded in the patients with polyposis and CRC (colorectal cancer). Increase in the number of sulphate reducing bacteria and production of H2S (a cytotoxic factor) and at the same time, decrease in the number of sulphide oxidizing bacteria, such as desulfovibrio, has been suggested as a probable etiological factor of CRD and polyposis[96,97]. In a separate research, increases in the levels of some probiotic bacteria, like Faecalibacterium, Fusobacterium, Roseburia andCoriobacteridae and a decrease in the level of enterobacteriaceae were observed within the gut of CRC afflicted patients[98].

Nowadays, researches regarding the effect(s) of gut microbial composition on psychology (mental health and mood) of an individual are progressively reported. The intestinal flora can influence development of brain cells and personal behaviors[99]. It has been proven that any alteration in the probiotic bacterial composition can directly affect the neurochemical secretions outside the gut. An intensified response in the hypothalamic-pituitary-adrenal axis to stresses, and at the same time, meaningful reductions in the production of brain-derived neurotrophic factors by the hippocampus and the cortex are proven through investigation on experimental animals[100,101]. Furthermore, a study on GF mice showed that the gut microbial flora affect the post-natal development of brain, control of brain motor and occurrence of an anxiety-like behavior by modulation of synaptophysin and PSD-95 (two critical glycoproteins involved in maturation of neuronal synapses)[102,103]. The gut microbial community might also affect the neuronal development and person’s behaviour by modulation neuronal transmitters such as acetylcholine, GABA (gamma-aminobutyric acid), melatonin, serotonin and histamines) within the intestinal tracts[102,103]. Based on the gut-brain communication hypothesis, it appears that gut microbial composition is closely associated with the psychiatric disorders such as depression[6]. Autism is the best known neural disorder associated with alteration of gut microbiota where the number of (spore forming) clostridia, specially Clostridium bolteae is meaningfully increase[104,105]. Based on this theory, it is easy to describe the reason of family involvement to the autism and the cause of its relapse after the treatment is cut[105]. Furthermore, it has been indicated that roughly 30% of the patients suffering of Major depressive disorder (MDD) are afflicted by irritable bowel syndrome (IBS)[106]. As it has been mentioned, IBS itself is caused partly by increase in the number of aerobic bacteria. It appears that the number of aerobic bacteria in the intestinal tract is critical and increase in the number of these bacteria can be associated with different physiological and psychological disorders. In addition to the case of MDD, it has been shown that the severity of neurological signs in fibromyalgia(FM) and Chronic Fatigue Syndrome (CFS) is directly associated with the abundance of these group of bacteria in the gut[107-110]. However, it is important to be clarified whether the psychological disorders are caused as a result of alteration in the composition of bacteria or inversely, the changes are a side effect of the diseases due to change in dietary regimen, physical inactivity or any changes in the level of secretion of digestive factors such as acids, bile salts and enzymes. It has been proven that the cytokines involved in depressive symptoms, like TNFα (tumour necrosis factor alpha)[111,112] and Il-1β (interleukin1-beta)[113] decrease the secretion of HCl in the stomach. On the other hand, it is necessary to be mentioned that the signs of fatigue were reduced in the patients suffering from CFS who took a tablet containing Lactobacillus casei strain Shirota (LcS)[100].

It also has been shown that composition and distribution of microbial flora in gastrointestinal tracts is directly associated with absorption of nutrients and establishment of malnutrition[11,68,69]. For instance, extra-proliferation of some bacteria, such as Bacteroides, Clostridia and Enterococci are attributed to several disorders such as megaloblastic anemia (due to absorption of vitamin B12)[114], steatorrhea (due to imbalance absorption of fatty acids and monoglycerides)[115], reduction of the gastric acidity and deficiency in digestive enzymes[116], obesity[117-119], bowel cancer[120] and allergic disease[121]. studies on malnourished children showed significant increases in the population of Campylobacteraceae, Helicobacteraceae, Bacteroidaceae and Porphyromonadaceae[122]. In contrast, the gut of healthy children was mostly enriched by Actinomycetales, Burkholderiales, Halobacteriales, Plantomycetales, Bifidobacteriales, Pseudomonadales, Enterobacteriales, Chloroflexales, Desulfovibrionales, Xanthomonadales, Lactobacillales, Rhizobiales, Planctomycetales and Clostridiales, in a diminishing order[122]. These changes are accomplished with several relapsing gastrointestinal infections, weight loss and growth impairment in the malnourished children[123-127]. Also, it has been indicated that majority of the microbial genes involved in obesity were derived from Firmicutes (25%) and Actinobacteria (75%), while the majority of genes involved in leans was mainly derived from Bacteroidetes (42%)[10]. Such these changes have been demonstrated in the microbiome of obese and lean twins[128] and researchers were able to transfer the obesity phenotype from obese mouse to lean animals[117].

A direct correlation between the resident microbial diversity of HBF (human baby flora) and host metabolic profiles has also indicated in several studies. For instance, it has been indicated that colonization of microorganism in the intestine leads to increase in the level of phenyl-containing amino acids. Colonization of Clostridium sporogenes in the gut is also associated with increase in the level of indole-3-propionic acid in host serum[102,129]. Furthermore, based on investigations on HBF inoculated mice, higher levels of myo-inositol and ethanolamine in the duodenum, higher concentration of betaine and taurine in the ileum, higher glutathione level in the jejunum and lower concentrations of myo-inositol and taurine in the colon were measured in compare to other regions of their gut[130]. The metabolic ability of microorganisms can affect the bioavailability of several medicines like simvastatin (used for decreasing the serum levels of cholesterol), salicylazosulfapyridine (administrated for ulcerative colitis), l-Dopa (for treatment of Parkinson) and digoxin (administrated for treatment of congestive heart failure)[131]. Simvastatin, for instance, down-regulate production of hepatic cholesterol through inhibition of the activity of HMG-COA (3-hydroxy-3-methylglutaryl coenzyme A). A Hydroxylation/dehydroxylation, methylation and beta-oxidative activity applied by intestinal microorganisms convert simvastatin into 2-hydroxyisovaleric acid and some other simple organic acids, leading to decrease the drug bioavailability[132].

CONCLUSION

Before application of metagenomic approaches for environmental samples, our knowledge was limited to some effects of microbial communities on that environment. However, metagenomic approaches and bioinformatic technologies enable researchers to find the exact microbial diversity and gene composition of an environment and to investigate effects of the genes on the environment. The outcomes of these researches, based on modulation of gut microbiome, are potentially applicable for therapeutic approaches, like antibiotic therapy, bacteriophage therapy and probiotic methods used not only for intestinal disorder also for some physiological abnormalities caused directly or indirectly by the gut microbiota[133-135]. Probiotic therapies, for instance, are nowadays proposed for several gut-related illnesses within gut or outside of the tracts.

For the first time, when Metchnikoff (1970) could link between the longevity of Bulgarian peasants with normal flora of their colon, he called these organisms as probiotics. It has been proved that the intestinal microbiota consisting of saccharolytic microorganisms, including lactobacillus sp., Bifidobacteria sp., E. coli, Enterococci, Bacillus sp., Bacteroides, Faecalibacterium, Propionibacterium and saccharomyces boulardii, increase our resistance against opportunistic infections due to alteration in the balance between microbial communities of intestinal tracts[136-138]. These microorganisms apply their probiotic activity based on metabolic excretion, immunomodulation and epigenetic modifications[139,140]. Some of the effects exerted by probiotic microorganisms include: (a) occupation of different parts of intestine and thus, prevention of colonization and stabilization of pathogenic microorganisms[141]; (b) production of antimicrobial compounds (such as bacteriocins, antibiotics, H2O2)[142,143]; (c) detoxification of many poisons and carcinogens[144]; (d) Regulation of immune responses to pathogens via up-regulation of anti-inflammatory modules and suppression of proinflammatory modules[145]; (e) stimulation of local immune system (cellular immune system or or sIgA)[146]; (f) production of degrading enzymes[147]; (g) attenuating the activity of many microbial enzymes involved in their metabolism (like nitroreductase, azoreductase, beta-glucosidase, beta-glucuronidase, ornithine decarboxylase and tryptophanase[148-150]; (h) attenuating the activity of many microbial enzyme involved in their virulence (eg, neuraminidase and mucinase)[151,152]; (i) reducing the level of polyamines, indol, ammonia, nitrate and nitrite in intestinal tracts[153-155]; (j) improvement the function of liver via stimulating secretion of bile salts[119]; (k) adjustment the blood pressure[156,157], (l) increase in metabolism of cholesterol[158,159]; (m) positive impacts on proliferation and differentiation of epithelial cells in the intestine[160]; and (n) improvement of intestinal barrier function[138].

It has been shown that Lactobacillus sp. and Bifidobacterium sp. play as anticancer factors in the gut. The anticancer activities are applied by stimulation of the immune system[161], inactivation of mutagenic agents (such as nitrosamine) inhibition of the activity of microbial enzymes present in feces (such as nitroreductase) and reduction in tumor cell proliferation (such as the inhibitory role of the cell wall of lactobacilli on cellular proliferation)[162,163]. Also, it has been indicated that probiotics improve digestive activity of intestine in several ways like production of the degrading enzymes for proteins, carbohydrates and fiber[147], improving the permeability of intestinal cells[164,165], reduction of the intolerance to foods, such as lactose[166], reducing of intestinal inflammation[167,168], treatment of intestinal disorders such as diarrhea and constipation[169,170], treatment of malnutrition via increase in absorption of minerals and production of different vitamins (such as family B, biotin, A, E, K and folic acid)[171,172].

Overall, presence and activity of probiotics in intestine is closely associated with human health and longevity. Consumption of certain foods such as yogurt and alcohol affect positively and negatively (respectively) on the intestinal flora bacteria. Consumption of special strains of Lactobacillus (or fermented milk) adjusts blood pressure via production of a tripeptide that inhibits the activity of Angiotensin; this enzyme functions in hypertension[156,157]. Also, it has been shown that consumption of yogurt for a week can reduce the level of cholesterol in blood via absorption of cholesterol as well as production and accumulation of hydroxyl-methyl-glutarate-coenzyme A (HMG-CoA) (an inhibitor of accumulation of HMG-COA reductase)[158,159]. Furthermore, several non-digestible food referred to as prebiotics such as inulin-containing oligosaccharides and fructooligosaccharides pass through the small intestine without any digestion and are able to activate selectively the bacterial duplication of bifidobacteria and lactobacilli in the large intestine[173,174].

REFERENCES

1 Riesenfeld C, Schloss P, Handelsman J. METAGENOMICS: Genomic Analysis of Microbial Communities. Annual Reviews of Genetics 2004; 38: 525–552

2 Desai N, Antonopoulos D, Gilbert JA, Glass EM, Meyer F. From genomics to metagenomics. Current Opinion in Biotechnology 2012; 23(1): 72-76

3 Warnecke F, Luginbühl P, Ivanova N, Ghassemian M, Richardson TH, Stege JT, Cayouette M, McHardy AC, Djordjevic G, Aboushadi N, Sorek R, Tringe SG, Podar M, Martin HG, Kunin V, Dalevi D, Madejska J, Kirton E, Platt D, Szeto E, Salamov A, Barry K, Mikhailova N, Kyrpides NC, Matson EG, Ottesen EA, Zhang X, Hernández M, Murillo C, Acosta LG, Rigoutsos I, Tamayo G, Green BD, Chang C, Rubin EM, Mathur EJ, Robertson DE, Hugenholtz P, Leadbetter JR. Metagenomic and functional analysis of hindgut microbiota of a wood-feeding higher termite. Nature 2007; 450(7169): 560-565

4 Hugenholtz P and Tyson G. Metagenomics. Nature 2008; 455(25): 481-483.

5 Neu J. Gastrointestinal maturation and feeding. in Seminars in perinatology 2006; Elsevier.

6 Collins SM and Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology 2009; 136(6): 2003-2014

7 Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science 2005; 308(5728): 1635-1638

8 Hattori M and Taylor TD. The human intestinal microbiome: a new frontier of human biology. DNA research 2009; 16(1): 1-12

9 Frank DN, Amand ALS, Feldman RA, Boedeker EC, Harpaz N, NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences 2007; 104(34): 13780-13785.

10 Tschöp MH, Hugenholtz P, Karp CL. Getting to the core of the gut microbiome. Nat Biotechnol 2009; 27(4): 344-346

11 Bourlioux P, Koletzko B, Guarner F, Braesco V. The intestine and its microflora are partners for the protection of the host: report on the Danone Symposium “The Intelligent Intestine,” held in Paris, June 14, 2002. The American journal of clinical nutrition 2003; 78(4): 675-683

12 Favier CF, Vaughan EE, De Vos WM, Akkermans ADL. Molecular monitoring of succession of bacterial communities in human neonates. Applied and Environmental Microbiology 2002; 68(1): 219-226

13 Zoetendal EG, Collier CT, Koike S, Mackie RI, Gaskins HR. Molecular ecological analysis of the gastrointestinal microbiota: a review. The Journal of nutrition 2004; 134(2): 465-472

14 Dominguez-Bello MG, Costello EK, 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. Proceedings of the National Academy of Sciences 2010; 107(26): 11971-11975

15 Grönlund MM, Lehtonen OP, Eerola E, Kero P. Fecal microflora in healthy infants born by different methods of delivery: permanent changes in intestinal flora after cesarean delivery. Journal of pediatric gastroenterology and nutrition 1999; 28(1): 19-25

16 Neu J, Douglas-Escobar M, Lopez M. Microbes and the developing gastrointestinal tract. Nutrition in Clinical Practice 2007; 22(2): 174-182

17 Yoshioka H, Iseki KI, Fujita K. Development and differences of intestinal flora in the neonatal period in breast-fed and bottle-fed infants. Pediatrics 1983; 72(3): 317-321

18 Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS biology 2007; 5(7): e177

19 Favier CF, de-Vos WM, Akkermans AD. Development of bacterial and bifidobacterial communities in feces of newborn babies. Anaerobe 2003; 9(5): 219-229

20 Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell 2006; 124(4): 837-848

21 Klaassens ES, de-Vos WM, Vaughan EE. Metaproteomics approach to study the functionality of the microbiota in the human infant gastrointestinal tract. Applied and environmental microbiology 2007; 73(4): 1388-1392

22 AL, PP, NW, AL. Jeffrey I. Human nutrition, the gut microbiome and the immune system. Nature 2011; 474(7351): 327-336

23 Slots J and Slots H. Bacterial and viral pathogens in saliva: disease relationship and infectious risk. Periodontology 2000, 2011; 55(1): 48-69

24 Chen T, Yu WH, Izard J, Baranova OV, Lakshmanan A, Dewhirst FE. The Human Oral Microbiome Database: a web accessible resource for investigating oral microbe taxonomic and genomic information. Database: the journal of biological databases and curation 2010; 2010.

25 Segata N, et al. Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biol 2012; 13(6): R42

26 Segata N, Haake SK, Mannon P, Lemon KP, Waldron L. Gevers D, Huttenhower C, Izard J. Microbial flora of the normal esophagus. Diseases of the esophagus: official journal of the International Society for Diseases of the Esophagus/ISDE 1998; 11(4): 248

27 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

28 Pei Z, Bini EJ, Yang L, Zhou M, Francois F, Blaser MJ. Bacterial biota in the human distal esophagus. Proceedings of the National Academy of Sciences 2004; 101(12): 4250-4255

29 Nossa CW, et al. Design of 16S rRNA gene primers for 454 pyrosequencing of the human foregut microbiome. World journal of gastroenterology: WJG 2010; 16(33): 4135

30 Pei Z, Yang L, Peek RM, Jr Levine SM, Pride DT, Blaser MJ. Bacterial biota in reflux esophagitis and Barrett’s esophagus. World journal of gastroenterology: WJG 2005; 11(46): 7277

31 Hu Y, He LH, Xiao D, Liu GD, Gu YX, Tao XX, Zhang JZ. Bacterial flora concurrent with Helicobacter pylori in the stomach of patients with upper gastrointestinal diseases. World journal of gastroenterology: WJG 2012; 18(11): 1257

32 Wang ZK and Yang YS. Upper gastrointestinal microbiota and digestive diseases. World journal of gastroenterology: WJG 2013; 19(10): 1541

33 Sanduleanu S, Jonkers D, De Bruïne A, Hameeteman W, Stockbrügger RW. Double gastric infection with Helicobacter pylori and non Helicobacter pylori bacteria during acid suppressive therapy: increase of pro inflammatory cytokines and development of atrophic gastritis. Alimentary pharmacology & therapeutics 2001; 15(8): 1163-1175

34 Tlaskalová-Hogenová H, Stěpánková R, Kozáková H, Hudcovic T, Vannucci L, Tučková L, Rossmann P, Hrnčíř T, Kverka M, Zákostelská Z, Klimešová K, Přibylová J, Bártová J, Sanchez D, Fundová P, Borovská D, Srůtková D, Zídek Z, Schwarzer M, Drastich P, Funda DP. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free and gnotobiotic animal models of human diseases. Cellular & molecular immunology 2011; 8(2): 110-120

35 Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK. Helicobacter pylori infection and the risk of gastric carcinoma. New England Journal of Medicine 1991; 325(16): 1127-1131

36 Watanabe T, et al. Helicobacter pylori infection induces gastric cancer in Mongolian gerbils. Gastroenterology 1998; 115(3): 642-648

37 Watanabe T, Tada M, Nagai H, Sasaki S, Nakao M. Analysis of the microflora in the stomach of Mongolian gerbils infected with Helicobacter pylori. Journal of gastroenterology and hepatology 2010; 25(s1): S11-S14

38 Chenoll E, Casinos B, Bataller E, Astals P, Echevarría J, Iglesias JR, Balbarie P, Ramón D, Genovés S. Novel probiotic Bifidobacterium bifidum CECT 7366 strain active against the pathogenic bacterium Helicobacter pylori. Applied and environmental microbiology 2011; 77(4): 1335-1343

39 Tong JL, Ran ZH, Shen J, Zhang CX, Xiao SD. Meta analysis: the effect of supplementation with probiotics on eradication rates and adverse events during Helicobacter pylori eradication therapy. Alimentary pharmacology & therapeutics 2007; 25(2): 155-168

40 Kamiji MM and de Oliveira RB. Non-antibiotic therapies for Helicobacter pylori infection. European journal of gastroenterology & hepatology 2005; 17(9): 973-981

41 Sun YQ, Monstein HJ, Nilsson LE, Petersson F, Borch K. Profiling and identification of eubacteria in the stomach of Mongolian gerbils with and without Helicobacter pylori infection. Helicobacter 2003; 8(2): 149-157

42 Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science 2006; 313(5790): 1126-1130

43 De La Cochetière, M.-F., et al., Effect of antibiotic therapy on human fecal microbiota and the relation to the development of Clostridium difficile. Microbial ecology 2008; 56(3): 395-402

44 De La Cochetière MF, Durand T, Lalande V, Petit JC, Potel G, Beaugerie L. Influence of anti-Helicobacter triple therapy with metronidazole, omeprazole and clarithromycin on intestinal microflora. Alimentary pharmacology & therapeutics 2001; 15(9): 1445-1452

45 Dethlefsen L, Eckburg PB, Bik EM, Relman DA. Assembly of the human intestinal microbiota. Trends in Ecology & Evolution 2006; 21(9): 517-523

46 Wang M, Ahrné 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

47 Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ, Hardcastle JD. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 1988; 29(8): 1035-1041

48 Jones BV, Begley M, Hill C, Gahan CGM, Marchesi JR. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proceedings of the National Academy of Sciences 2008; 105(36): 13580-13585

49 Hao WL, Lee YK. Microflora of the gastrointestinal tract, in Public Health Microbiology. 2004, Springer. p. 491-502.

50 Neu J. Gastrointestinal maturation and feeding. Seminars in perinatology 2006; 30(2): 77-80

51 Swidsinski A, Loening-Baucke V, Vaneechoutte M, Doerffel Y. Active Crohn's disease and ulcerative colitis can be specifically diagnosed and monitored based on the biostructure of the fecal flora. Inflammatory bowel diseases 2008; 14(2): 147-161

52 Isaacs K and Herfarth H. Role of probiotic therapy in IBD. Inflammatory bowel diseases 2008; 14(11): 1597-1605

53 Wang X, Heazlewood SP, Krause DO, Florin TH. Molecular characterization of the microbial species that colonize human ileal and colonic mucosa by using 16S rDNA sequence analysis. Journal of applied microbiology 2003; 95(3): 508-520

54 Stevens CE, Hume ID. Contributions of microbes in vertebrate gastrointestinal tract to production and conservation of nutrients. Physiological Reviews 1998; 78(2): 393-427

55 Xu J, Gordon JI. Honor thy symbionts. Proceedings of the National Academy of Sciences 2003; 100(18): 10452-10459

56 Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE. Metagenomic analysis of the human distal gut microbiome. science 2006; 312(5778): 1355-1359

57 Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knigh R. Bacterial community variation in human body habitats across space and time. Science 2009; 326(5960): 1694-1697

58 Kinross JM, Darzi AW, Nicholson JK. Gut microbiome-host interactions in health and disease. Genome Med 2011; 3(3): 14.

59 Ott SJ, Kühbacher T, Musfeldt M, Rosenstiel P, Hellmig S, Rehman A, Drews O, Weichert W, Timmis KN, Schreiber S. Fungi and inflammatory bowel diseases: alterations of composition and diversity. Scandinavian journal of gastroenterology 2008; 43(7): 831-841

60 Scanlan PD and Marchesi JR. Micro-eukaryotic diversity of the human distal gut microbiota: qualitative assessment using culture-dependent and-independent analysis of faeces. The ISME journal 2008; 2(12): 1183-1193

61 Reyes A, Haynes M, Hanson N, Angly FE,Heath AC, Rohwer F, Gordon JI. Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature 2010; 466(7304): 334-338

62 Scanlan P and Marchesi J. Micro-eukaryotic diversity of the human distal gut microbiota: qualitative assessment using culture-dependent and -independent analysis of faeces. International Society for Microbial Ecology 2008; 2: 1183–1193

63 Pritt BS, Clark CG. Amebiasis. in Mayo Clinic Proceedings 2008; Elsevier.

64 Rajilić-Stojanović M, Smidt H, De Vos WM. Diversity of the human gastrointestinal tract microbiota revisited. Environmental microbiology 2007; 9(9): 2125-2136

65 Mihajlovski A, Alric M, Brugère JF. A putative new order of methanogenic Archaea inhabiting the human gut, as revealed by molecular analyses of the gene. Research in microbiology 2008; 159(7): 516-521

66 Stecher B and Hardt WD. The role of microbiota in infectious disease. Trends in microbiology 2008; 16(3): 107-114

67 Savage DC. Microbial ecology of the gastrointestinal tract. Annual Reviews in Microbiology 1977; 31(1): 107-133

68 Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proceedings of the National Academy of Sciences 2002; 99(24): 15451-15455

69 Todar K. Todar's online textbook of bacteriology. 2006: University of Wisconsin-Madison Department of Bacteriology.

70 Wilson I, Nicholson J. The role of gut microbiota in drug response. Current pharmaceutical design 2009; 15(13): 1519-1523

71 Jia W, Li H, Zhao L, Nicholson JK. Gut microbiota: a potential new territory for drug targeting. Nature Reviews Drug Discovery 2008; 7(2): 123-129

72 Mason KL, Huffnagle GB, Noverr MC, Kao JY. Overview of gut immunology, in GI Microbiota and Regulation of the Immune System. 2008; Springer. p. 1-14.

73 Yamanaka T, Helgeland L, Farstad IN, Fukushima H, Midtved T, Brandtzaeg P. Microbial colonization drives lymphocyte accumulation and differentiation in the follicle-associated epithelium of Peyer’s patches. The Journal of Immunology 2003; 170(2): 816-822

74 Eckmann L, Stenson WF, Savidge TC, Lowe DC, Barrett KE, Fierer J, Smith JR, Kagnoff MF. Role of intestinal epithelial cells in the host secretory response to infection by invasive bacteria. Bacterial entry induces epithelial prostaglandin h synthase-2 expression and prostaglandin E2 and F2alpha production. Journal of Clinical Investigation 1997; 100(2): 296

75 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(2): 229-241

76 Artis D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nature Reviews Immunology 2008; 8(6): 411-420

77 Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, Xavier RJ, Teixeira MM, Mackay CR. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461(7268): 1282-1286

78 Vinolo MAR, Rodriguesemail HG, Nachbaremail RT, Curiemail R. Regulation of inflammation by short chain fatty acids. Nutrients, 2011. 3(10): p. 858-876.

79 Gracey M. Intestinal microflora and bacterial overgrowth in early life. Journal of Pediatric Gastroenterology and Nutrition 1982; 1(1): 13-22

80 Hooper LV, Stappenbeck TS, Hong CV, Gordon JI. Angiogenins: a new class of microbicidal proteins involved in innate immunity. Nature immunology 2003; 4(3): 269-273

81 Lakhdari O, Cultrone A, Tap J, Gloux K, Bernard F, Ehrlich SD, Lefèvre F, Doré J, Blottière HM. Functional metagenomics: a high throughput screening method to decipher microbiota-driven NF-κB modulation in the human gut. PLoS One 2010; 5(9): e13092

82 Hayden MS and Ghosh S. Shared principles in NF-κB signaling. Cell 2008; 132(3): 344-362

83 Olefsky JM. IKK: A Bridge between Obesity and Inflammation. Cell 2009; 138(5): 834-836

84 Eckmann L, Nebelsiek T, Fingerle AA, Dann SM, Mages J, Lang R, Robine S, Kagnoff MF, Schmid RM, Karin M, Arkan MC, Greten FR. Opposing functions of IKKβ during acute and chronic intestinal inflammation. Proceedings of the National Academy of Sciences 2008; 105(39): 15058-15063

85 Baumgart DC, Carding SR. Inflammatory bowel disease: cause and immunobiology. The Lancet 2007; 369(9573): 1627-1640

86 Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology 1998; 115(1): 182-205

87 Macpherson AJ and Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nature Reviews Immunology 2004; 4(6): 478-485

88 Björkstén B. Effects of intestinal microflora and the environment on the development of asthma and allergy. in Springer seminars in immunopathology 2004; Springer.

89 Björkstén B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal microflora during the first year of life. Journal of Allergy and Clinical Immunology 2001; 108(4): 516-520

90 Nadal I, Donat E, Ribes-Koninckx C, Calabuig M, Sanz Y. Imbalance in the composition of the duodenal microbiota of children with coeliac disease. Journal of Medical Microbiology 2007; 56(12): 1669-1674

91 Nistal E, Caminero A, Herrán AR, Arias L, Vivas S, de Morales JM, Calleja S, de Miera LE, Arroyo P, Casqueiro J. Differences of small intestinal bacteria populations in adults and children with/without celiac disease: effect of age, gluten diet, and disease. Inflammatory Bowel Diseases 2012; 18(4): 649-656

92 Sokol H and Seksik P. The intestinal microbiota in inflammatory bowel diseases: time to connect with the host. Current opinion in gastroenterology 2010; 26(4): 327-331

93 Franchimont D, Vermeire S, El Housni H, Pierik M, Van Steen K, Gustot T, Quertinmont E, Abramowicz M, Van Gossum A, Devière J, Rutgeerts P. Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn’s disease and ulcerative colitis. Gut 2004; 53(7): 987-992

94 Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB, Bousvaros A, Korzenik J, Sands BE, Xavier RJ, Huttenhower C. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012; 13(9): R79

95 Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, Housseau F, Pardoll DM, Sears CL. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nature medicine 2009; 15(9): 1016-1022

96 Balamurugan R, Rajendiran E, George S, Samuel GV, Ramakrishna BS. Real-time polymerase chain reaction quantification of specific butyrate-producing bacteria, Desulfovibrio and Enterococcus faecalis in the feces of patients with colorectal cancer. Journal of gastroenterology and hepatology 2008; 23(8pt1): 1298-1303

97 Scanlan PD, Shanahan F, Clune Y, Collins JK, O'Sullivan GC, O'Riordan M, Holmes E, Wang Y, Marchesi JR. Culture-independent analysis of the gut microbiota in colorectal cancer and polyposis. Environmental microbiology 2008; 10(3): 789-798

98 Marchesi JR, Dutilh BE, Hall N, Peters WHM, Roelofs R, Boleij A, Tjalsma H. Towards the human colorectal cancer microbiome. PloS one 2011; 6(5): e20447

99 Heijtza RD, Wang S, Anuard F, Qiana Y, Björkholm B, Samuelsson A, Hibber ML, Forssberg H, Pettersson S. Normal gut microbiota modulates brain development and behavior. Proceedings of the National Academy of Sciences 2011; 108(7): 3047-3052

100 Rao AV, Bested AC, Beaulne TM, Katzman MA, Iorio C, Berardi JM, Logan AC. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathogens 2009; 1(1): 1-6

101 Bercik P, Verdu EF, Foster JA, Macri J, Potter M, Huang X, Malinowski P, Jackson W, Blennerhassett P, Neufeld KA, Lu J, Khan WI, Corthesy-Theulaz I, Cherbut C, Bergonzelli GE, Collins SM. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology 2010; 139(6): 2102-2112 e1

102 Wikoffa WR, Anforab AT, Liub J, Schultz PG, Lesley SA, Petersb EC, Siuzdaka G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proceedings of the National Academy of Sciences 2009; 106(10): 3698-3703

103 Uribe A, Alam M, Johansson O, Midtvedt T, Theodorsson E. Microflora modulates endocrine cells in the gastrointestinal mucosa of the rat. Gastroenterology 1994; 107(5): 1259-1269

104 MRT Parracho HMRT, Bingham MO, Gibson GR, McCartney AL. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. Journal of Medical Microbiology 2005; 54(10): 987-991

105 Finegold SM. Therapy and epidemiology of autism–clostridial spores as key elements. Medical hypotheses 2008; 70(3): 508-511

106 Logan AC and Katzman M. Major depressive disorder: probiotics may be an adjuvant therapy. Medical hypotheses 2005; 64(3): 533-538

107 Lakhan SE and Kirchgessner A. Gut inflammation in chronic fatigue syndrome. Nutr Metab (Lond) 2010; 7: 79

108 Logan AC, Venket Rao A, Irani D. Chronic fatigue syndrome: lactic acid bacteria may be of therapeutic value. Medical hypotheses 2003; 60(6): 915-923

109 Nicolson GL, Nasralla MY, Franco AR, Nicolson NL, Erwin R, Ngwenya R, Paul A. Berns PA.Diagnosis and Integrative Treatment of Intracellular Bacterial Infections in Chronic Fatigue and Fibromyalgia Syndromes, persian Gulf War Illness, Rheumatoid Arthritis, and Other Chronic Illnesses. Clinical Practice of Alternative Medicine 2000; 1: 92-106

110 Pimentel M, Wallace D, Hallegua D, Chow E, Kong Y, Park S, Lin HC. A link between irritable bowel syndrome and fibromyalgia may be related to findings on lactulose breath testing. Annals of the rheumatic diseases 2004; 63(4): 450-452

111 Suarez EC, Krishnan RR, Lewis JG. The relation of severity of depressive symptoms to monocyte-associated proinflammatory cytokines and chemokines in apparently healthy men. Psychosomatic medicine 2003; 65(3): 362-368

112 Suarez EC, Lewis JG, Krishnan RR, Young KH. Enhanced expression of cytokines and chemokines by blood monocytes to in vitro lipopolysaccharide stimulation are associated with hostility and severity of depressive symptoms in healthy women. Psychoneuroendocrinology 2004; 29(9): 1119-1128

113 El-Omar E., The importance of interleukin 1β inHelicobacter pylori associated disease. Gut 2001; 48(6): 743-747

114 Singh VV and Toskes PP. Small bowel bacterial overgrowth: presentation, diagnosis, and treatment. Current treatment options in gastroenterology 2004; 7(1): 19-28

115 Donaldson R. Studies on the pathogenesis of steatorrhea in the blind loop syndrome. Journal of Clinical Investigation 1965; 44(11): 1815

116 Sartor R. Review article: Role of the enteric microflora in the pathogenesis of intestinal inflammation and arthritis. Alimentary pharmacology & therapeutics 1997; 11(s3): 17-23

117 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

118 Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature 2006; 444(7122): 1022-1023

119 Loguercio C, Federico A, Tuccillo C, Terracciano F, D'Auria MV, De Simone C, Del Vecchio Blanco C. Beneficial effects of a probiotic VSL# 3 on parameters of liver dysfunction in chronic liver diseases. Journal of clinical gastroenterology 2005; 39(6): 540-543

120 Lampe JW. The Human Microbiome Project: getting to the guts of the matter in cancer epidemiology. Cancer Epidemiology Biomarkers & Prevention 2008; 17(10): 2523-2524

121 Vassallo M and Walker W. Neonatal microbial flora and disease outcome. 2008.

122 Gupta SS et al. Metagenome of the gut of a malnourished child. Gut Pathog 2011; 3(1): 7

123 Windle HJ, Kelleher D, Crabtree JE. Childhood Helicobacter pylori infection and growth impairment in developing countries: a vicious cycle? Pediatrics 2007; 119(3): e754-e759

124 Clemens J, Albert MJ, Rao M, Huda S, Qadri F, Van Loon FP, Pradhan B, Naficy A, Banik A. Sociodemographic, hygienic and nutritional correlates of Helicobacter pylori infection of young Bangladeshi children. The Pediatric infectious disease journal 1996; 15(12): 1113-1118.

125 Bhan MK, Bahl R, Sazawal S, Sinha A, Kumar R, Mahalanabis D, Clemens JD. Association between Helicobacter pylori infection and increased risk of typhoid fever. Journal of Infectious Diseases 2002; 186(12): 1857-1860

126 Patel P, Mendall MA, Khulusi S, Northfield TC, Strachan DP. Helicobacter pylori infection in childhood: risk factors and effect on growth. Bmj 1994; 309(6962): 1119-1123

127 DiBaise JK, Zhang H, Crowell MD, Krajmalnik-Brown R, Decker GA, Rittmann BE. Gut microbiota and its possible relationship with obesity. in Mayo Clinic Proceedings. 2008. Elsevier.

128 Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature 2008; 457(7228): 480-484

129 Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nature Reviews Microbiology 2005; 3(5): 431-438

130 Martin FP, Wang Y, Yap IK, Sprenger N, Lindon JC, Rezzi S, Kochhar S, Holmes E, Nicholson JK. Topographical variation in murine intestinal metabolic profiles in relation to microbiome speciation and functional ecological activity. Journal of Proteome Research 2009; 8(7): 3464-3474

131 Sousa T, Paterson R, Moore V, Carlsson A, Abrahamsson B, Basit AW. The gastrointestinal microbiota as a site for the biotransformation of drugs. International journal of pharmaceutics 2008; 363(1): 1-25

132 Aura AM, Mattila I, Hyötyläinen T, Gopalacharyulu P, Bounsaythip C, Orešič M, Oksman-Caldentey KM. Drug metabolome of the Simvastatin formed by human intestinal microbiota in vitro. Molecular BioSystems 2011; 7(2): 437-446

133 Borody TJ, Warren EF, Leis S, Surace R, Ashman O. Treatment of ulcerative colitis using fecal bacteriotherapy. Journal of clinical gastroenterology 2003; 37(1): 42-47

134 Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, Bazzocchi G, Poggioli G, Miglioli M, Campieri M. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology 2000; 119(2): 305-309

135 Chibani-Chennoufi S, Sidoti J, Bruttin A, Kutter E, Sarker S, Brüssow H. In vitro and in vivo bacteriolytic activities of Escherichia coli phages: implications for phage therapy. Antimicrobial agents and chemotherapy 2004; 48(7): 2558-2569

136 Vieira LQ, dos Santos LM, Neumann E, da Silva AP, Moura LN, Nicoli JR. Probiotics protect mice against experimental infections. Journal of clinical gastroenterology 2008; 42: S168-S169.

137 Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M, Ohno H. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011; 469(7331): 543-547

138 Preidis GA and Versalovic J. Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology 2009; 136(6): 2015-2031

139 Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Zhang M, Wei H, Chen Y, Lu H, Zuo J, Su M, Qiu Y, Wei Jia W, Xiao C, Smith LM, Yang S, Holmes E, Tang H, Zhao G, Nicholson JK, Li L, Zhao L. Symbiotic gut microbes modulate human metabolic phenotypes. Proceedings of the National Academy of Sciences 2008; 105(6): 2117-2122

140 Licciardi PV, Wong S, Tang ML, Karagiannis TC. Epigenome targeting by probiotic metabolites. Gut Pathog 2010; 2(1): 24

141 Salminen S, Isolauri E, Salminen E. Clinical uses of probiotics for stabilizing the gut mucosal barrier: successful strains and future challenges. Antonie Van Leeuwenhoek 1996; 70(2-4): 347-358

142 Pinchuk IV, Bressollier P, Verneuil B, Fenet B, Sorokulova IB, Mégraud F, Urdaci MC. In Vitro Anti-Helicobacter pyloriActivity of the Probiotic Strain Bacillus subtilis 3 Is Due to Secretion of Antibiotics. Antimicrobial agents and chemotherapy 2001; 45(11): 3156-3161

143 Ouwehand AC and Vesterlund S. Antimicrobial components from lactic acid bacteria. Lactic acid bacteria microbiological and functional aspects, 2004. 3

144 Ziemer CJ and Gibson GR. An overview of probiotics, prebiotics and synbiotics in the functional food concept: perspectives and future strategies. International Dairy Journal 1998; 8(5-6): 473-479

145 Tlaskalová-Hogenová H, Stepánková R, Hudcovic T, Tucková L, Cukrowska B, Lodinová-Zádníková R, Kozáková H, Rossmann P, Bártová J, Sokol D, Funda DP, Borovská D, Reháková Z, Sinkora J, Hofman J, Drastich P, Kokesová A. Commensal bacteria (normal microflora), mucosal immunity and chronic inflammatory and autoimmune diseases. Immunology letters 2004; 93(2): 97-108

146 Erickson KL, Hubbard NE. Probiotic immunomodulation in health and disease. The Journal of nutrition 2000; 130(2): 403S-409S

147 Pharmaceutiques UdL. Dietary modulation of the human colonie microbiota: introducing the concept of prebiotics. Journal of Nutrition 1995; 125: 1401-1412

148 De Preter V, Raemen H, Cloetens L, Houben E, Rutgeerts P, Verbeke K. Effect of dietary intervention with different pre-and probiotics on intestinal bacterial enzyme activities. European journal of clinical nutrition 2007; 62(2): 225-231

149 Bomba A, Nemcová R, Gancarcíková S, Herich R, Guba P, Mudronová D. Improvement of the probiotic effect of micro-organisms by their combination with maltodextrins, fructo-oligosaccharides and polyunsaturated fatty acids. British Journal of Nutrition 2002; 88(S1): S95-S99

150 O’Bryan CA, Crandall PG, Lee SO, Ricke SC. The Role of Prebiotics and Probiotics in Human Health. J Prob Health 2013; 1(108): 2

151 Szabady RL. Secreted Mucinases of Enteric Bacterial Pathogens: Modulators of Host Defense. 2008: ProQuest

152 Wilson BA, Thomas SM, Ho M. The human vaginal microbiome, in Metagenomics of the Human Body. 2011, Springer 91-115

153 Rowland RH, Ian R. Metabolic activities of the gut microflora in relation to cancer. Microbial Ecology in Health and Disease 2000; 12(2): 179-185

154 Sobko T, Reinders CI, Jansson E, Norin E, Midtvedt T, Lundberg JO. Gastrointestinal bacteria generate nitric oxide from nitrate and nitrite. Nitric Oxide 2005; 13(4): 272-278

155 Linsalata M, Russo F, Berloco P, Caruso ML, Matteo GD, Cifone MG, Simone CD, Ierardi E, Di Leo A. The Influence of Lactobacillus brevis on Ornithine Decarboxylase Activity and Polyamine Profiles in Helicobacter pylori-Infected Gastric Mucosa. Helicobacter 2004; 9(2): 165-172

156 Ong L, Shah NP. Release and identification of angiotensin-converting enzyme-inhibitory peptides as influenced by ripening temperatures and probiotic adjuncts in Cheddar cheeses. LWT-Food Science and Technology 2008; 41(9): 1555-1566

157 Quirós A, Hernández-Ledesma B, Ramos M, Amigo L, Recio I. Angiotensin-converting enzyme inhibitory activity of peptides derived from caprine kefir. Journal of dairy science 2005; 88(10): 3480-3487

158 Liong MT. Probiotics: a critical review of their potential role as antihypertensives, immune modulators, hypocholesterolemics, and perimenopausal treatments. Nutrition reviews 2007; 65(7): 316-328

159 Endo T, Nakano M, Shimizu S, Fukushima M, Miyoshi S. Effects of a probiotic on the lipid metabolism of cocks fed on a cholesterol-enriched diet. Bioscience, biotechnology, and biochemistry 1999; 63(9): 1569-1575

160 Becker S, Oelschlaeger TA, Wullaert A, Pasparakis M, Wehkamp J, Stange EF, Gersemann M. Bacteria Regulate Intestinal Epithelial Cell Differentiation Factors Both In Vitro and In Vivo. PloS one 2013; 8(2): e55620

161 Perdigon G, Alvarez S, Rachid M, Agüero G, Gobbato N. Immune system stimulation by probiotics. Journal of dairy science 1995; 78(7): 1597-1606

162 Yan F, Cao H, Cover TL, Whitehead R, Washington MK, Polk DB. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007; 132(2): 562-575

163 Hirayama K and Rafter J. The role of probiotic bacteria in cancer prevention. Microbes and infection 2000; 2(6): 681-686

164 Madsen K, Cornish A, Soper P, McKaigney C, Jijon H, Yachimec C, Doyle J, Jewell L, De Simone C. Probiotic bacteria enhance murine and human intestinal epithelial barrier function. Gastroenterology 2001; 121(3): 580-591

165 Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ, Wells JM, Roy NC. Regulation of tight junction permeability by intestinal bacteria and dietary components. The Journal of nutrition 2011; 141(5): 769-776

166 Lourens-Hattingh A, Viljoen BC. Yogurt as probiotic carrier food. International Dairy Journal 2001; 11(1): 1-17

167 Isolauri E, Kirjavainen P, Salminen S. Probiotics: a role in the treatment of intestinal infection and inflammation? Gut 2002; 50(suppl 3): iii54-iii59

168 Marteau PR, de Vrese M, Cellier CJ, Schrezenmeir J. Protection from gastrointestinal diseases with the use of probiotics. The American journal of clinical nutrition 2001; 73(2): 430s-436s

169 Whorwell PJ, Altringer L, Morel J, Bond Y, Charbonneau D, O'Mahony L, Kiely B, Shanahan F, Quigley EM. Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. The American journal of gastroenterology 2006; 101(7): 1581-1590

170 Yan F, Polk DB. Probiotics as functional food in the treatment of diarrhea. Current Opinion in Clinical Nutrition & Metabolic Care 2006; 9(6): 717-721

171 Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria. Nutrients 2011; 3(1): 118-134

172 Farnworth ER. Kefir–a complex probiotic. Food Science and Technology Bulletin: Fu 2006; 2(1): 1-17

173 Kleessen B, Sykura B, Zunft HJ, Blaut M. Effects of inulin and lactose on fecal microflora, microbial activity, and bowel habit in elderly constipated persons. The American journal of clinical nutrition 1997; 65(5): 1397-1402

174 de Vrese M, Schrezenmeir J. Probiotics, prebiotics, and synbiotics, in Food Biotechnology. 2008, Springer. 1-66


Peer reviewers: I. Michael Leitman, MD, Chief Of General Surgery, Albert Einstein College Of Medicine-Beth Israel Medical Center, 10 Union Square East, 2m, New York, 10003, the United States; Dana Telem, Assistant Professor of Surgery , Associate Fellowship Director, Stony Brook University Medical Center, Department of Surgery, Health Sciences Center T18-040, Stony Brook Medicine, Stony Brook, NY 11794-8191, the United States; Wen Xie Xu, Professor, Department of Physiology, Shanghai Jiaotong University School of Medicine, 328 room, Wenxuan Medical Building, 800 Dongchuan Rd, Shanghai, 200240, China.

Refbacks

  • There are currently no refbacks.


Creative Commons License
This work is licensed under a Creative Commons Attribution 3.0 License.