Home » Endocrinology » IJCE-ID23.php International Journal of Clinical Endocrinology - ISSN: 2640-5709


Mini-Review

Microbial Endocrinology in Microbiology: A Mini-Review

Michael AB. Naafs*

Dutch Internist Endocrinologist, Health Consultant at Naafs, International Health Consultancy, Rhodoslaan 20, 7577KN, Oldenzaal, The Netherlands

*Address for Correspondence: Michael AB. Naafs, Dutch Internist Endocrinologist, Health Consultant at Naafs, International Health Consultancy, Rhodoslaan 20, 7577KN, Oldenzaal, The Netherlands, Tel:+31-681-589-079 ; ORCID: https://orcid.org/0000-0002-6788-9399, E-mail: naafs.healthconsultancy@gmail.com; michael.naafs@hotmail.com

Submitted: 22 January 2018; Approved: 23 January 2018; Published: 25 January 2018

Citation this article: Naafs MAB. Microbial Endocrinology in Microbiology: A Mini-Review. Int J Clin Endocrinol. 2018;2(1): 004-010.

Copyright: © 2018 Naafs MAB. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited

Download Fulltext PDF

Microbial endocrinology represents the intersection of two seemingly desparate fields, microbiology and neurobiology. Current research developments in intestinal microbiota and microbial endocrinology are considered. Its relationship with behavior, metabolism and a variety of diseases are discussed. Pathogenesis, pathophysiology and possible therapeutic applications to the intestinal microbiome communities are reviewed.

Introduction

Microbial endocrinology represents the intersection of two seemingly desperate fields, microbiology and neurobiology. This is based on the shared presence of neurochemicals that are exactly the same in the host as well as in the microorganism. Production of neurochemicals by microorganisms most often employs the same biosynthetic pathways as those utilized by the host, indicating that acquisition of host neurochemical-based signalling system in the host may have been acquired due to lateral gene transfer from microorganisms. Current perceptions of how stress influences the outcome of infections focus upon the immunology and leave the microbe largely as a bystander.

Stress by the central nervous system leads to release a variety of hormones, neurochemicals and neuropeptides, which can directly affect immune function, usually resulting in impairment [1]. Nearly all immune cell classes possess receptors for the stress-related neurohormones adrenaneline and noradrenaline [2]. The ability of bacterial pathogens to influence behavior has been recognized for decades, most notably bacteria that invade the nervous system. The term microbial endocrinolgy and the concept of the “gut microbiome-brain axis” developed in the early 1990’s. Since their introduction both concepts have been the subject of growing investigations. In this mini-review pathogenesis, pathophysiology and the therapeutic applications of the microbial endocrinology in microbiology will be discussed.

Pathogenesis

Although most microbial endocrinology studies have focused on the interaction of gut bacteria with the fight and flight catecholamines adrenaline, noradrenaline and dopamine, it is important to realize that bacteria and fungi can recognize a surprising number of eukaryotic hormones and other signals [3]. Structurally, the catecholamine stress hormone family are a group of widely acting acting effector compounds derived from tyrosine and other dietary aminoacids. They chemically comprise a benzene ring with two adjacent hydroxyl groups and an opposing amino side chain which contributes to receptor specificity [4]. The catecholamines use the second messenger adenylate cyclase system to exert their downstream effects after receptor binding [5].

The synthesis pathway for catecholamines begins with dietary L-dopa, which is enzymatically converted into dopamine, norepinephrine and finally adrenaline. Noradrenergic and dopaminergic receptors containing nerve terminals are widely distributed within the mammalian body, including the GI tract where they are components of the enteric nervous system. Further research of microbial endocrinolgy discovered hormone receptors in microorganisms and it was hypothesized that they represent a form of intercellular communication [8]. Pathogenic neurotoxins such as neurotoxin 6-hydroxydopamine were shown to alter norepinephrine levels in mice presenting the bi-directional nature of the host-microbe interaction [9]. Iyer et al. [10] showed that many enzymes involved in host hormone metabolism (including epinephrine, norepinephrine, dopamine, serotonine, melatonin etc.) might have evolved horizontal gene transfer from bacteria.

More clues to the existence of crosstalk between bacteria and the endocrine system came from the discovery of inter-kingdom, including the hormonal communication between microorganisms and their hosts [11]. It appeared from the initial observation that bacteria perform Quorum Sensing (QS), communication based on producing and sensing Autoinducer (AI) molecules. These AI molecules are hormone-like elements that regulate functions, including bacterial growth, motility and virulence [12]. In addition, to affecting bacteria, these signals can modulate host cell signal transduction. Some AI molecules have crosstalks with host hormones for activating signalling pathways [13].

Host hormones also affect bacterial gene expression which in turn can have consequences on their hosts [14]. For example, catecholamines enhance bacterial attachment to host tissues [12]. Quorum sensing is enhanced by catecholamines, but inhibited by the human sex hormones estriol and estradiol [15].

Pathophysiology

Endocrine effects of bacteria influence a variety of host responses including, behavior, metabolism and appetite and immune response. Much of the advances in this field in its infancy have been made through experiments, using germ-free animals, as well as experiments using probiotics (specific microbes thought to be beneficial to the host) and prebiotics (non-digestible carbohydrates that act as food for probiotics), together with advances in sequencing and bioinformatics platforms.

Behavior

The ability of pathogens to influence host behavior has been known for long times. An example is Toxoplasma gondii infections of rodents, that result in such a profound decrease in anxiety, that infected animals no longer showed fear of feline predators [15]. Humans suffering from inflammatory bowel diseases, which are characterized by disturbed enteric microbial diversity, demonstrated poorer emotional function such as anxiety and depression [16]. Release of host immune factors, such as cytokines and inflammatory mediators, that have neuronal targets, both within the CNS and the Enteric Nervous System (ENS) are believed to be involved [17].

The first study that demonstrated the ability of a bacterium within the gut to influence behavior was shown in a series of studies using Campylobacter jejuni in mice [18]. In this series of studies, a low per oral dose of C. jejuni was able to induce anxiety-like behavior in mice through a vagal-mediated pathway in the absence of immune activation [19]. Within the gut neuronal projections from the ENS can innervate the entire length of the microvilli [20,21]. Coupled with the presence of a myriad of cells within the gastrointestinal tract, such as enterochromaffin cells and luminal epithelial chemosensors, there is a host of information, that can be shared with the CNS, such as he brain [22]. Neufeld et al. showed that excitability of gut sensory neurons located within the myenteric plexus of the ENS, isolated from jejunal segments of the intestine, relied on the presence of the normal commensal microbiota for proper functioning [23].

Synthesis of benzodiazepine receptor ligands by gut bacteria can contribute to the development of encephalopathy that can accompany fulminant hepatic failure by accumulating in the brain and enhancing GABA ( gamma-aminobutyric acid ) inhibitory neurotransmission system. Subsequent reports identified the neural substrates both within the brain and vagal-mediated gut-to-brain pathway [19]. For example, the ability of cetain probiotic bacteria such as Lactobacillus rhamnosus to infkuence emotion behavior in mice has been shown to be mediated via GABA receptors [25]. Changes in diet such as feeding of meat, which can dramatically alter the composition of the microbiome, have been shown to improve memory and learning in rodents [26]. It should not be surprising that the intestinal microbiome plays a critical role in the development of the brain itself from the time of birth [27].

Appetite and metabolism

A classic role of the gut microbiota is in digesting a variety of carbohydrates and fermenting them into short-chain fatty acides (SCFAs). Germ-free (GF) mice have different metabolic profiles than conventionally raised mice, including low concentrations of SCFAs, hepatic triacylglycerol and glucose. Subtherapeutic doses of antibiotics, which do not eliminate the gut microbial community, but rather cause significant changes in the composition, lead to increased levels of SCFAs and to weight gain in mice [28]. These metabolic effects of the microbiome may further affect hormone production. For instance SCFAs have been shown to stimulate release of 5-HT (5-hydroxytryptamine or serotonine) and the peptide YY, a hormone released after feeding involved in appetite reduction and slowing gut motility [29,30]. Although a lot of neuropeptides that have a role in controlling appetite and regulating metabolism could be affected by the gut microbiome, this is until now more speculative than evidence-based.

Potential candidates are; alpha-MSH (melanocyte-stimulating hormone, neuropeptide YY, agouti-related protein, ghrelin, leptin, insulin and others. Somatostatin, which suppresses the release of the GI and pancreatic hormones is of interest too [31]. Several pieces of evidence link the microbiota function to leptin levels. Use of antibiotica (vancomycin) in rats leads to a a dramatic decline (38%) in crculating leptin levels [32]. Several bacteria genera (e.g., Allobaculum, Clostridium, Bacteroides and Prevotella) correlate negatively with leptin levels, while others(e.g., Mucispirillum, Lactococcus, Bifidobacterium ) correlate positively with circulating leptin concentrations in mice These correlations may stem from bacteria affecting hormone levels, or vice versa. One proposed mechanism is that diet composition may impact leptin concentrations which, in turn, may change the microbial community composition through inflammatory and or regulation of mucus production [33,34]. Rajala et al. [35] showed that leptin might also influence the gut microbiota independently of diet. Another model proposes that Lactobacillus plantarum specifically suppresses leptin by reducing adipocyte cell size in white tissue fat [32,36]. This fits the finding that the use of the probiotic L. plantarum in a group of human smokers reduced their serum leptin levels [37]. Leptin is involved in appetite inhibition, metabolism and behavior and therefore its possible interconnections with bacteria could be of great interest.

Grhelin, another appetite-regulating hormone is negatively correlated with the abundance of Bifidbacterium, Lactobacillus and B, coccoides-Eubacterium rectale group, and positively correlated with a number of Bacteroides and Prevotella species [34]. Intake of oligofructose (a prebiotic that promotes growth of Bifidobacterium and Lactobacillus) decreases secretion of grhelin in obese humans [38].

Insulin may provide another link between the microbiome and hormones. Significant variations in microbiome composition have been observed in diabetic patients compared to healthy controls. Certain bacterial species have been positively or negatively correlated with insulin levels [39,40]. Transfer of the intestinal microbiota ( including butyrate producing microbiota) from lean donors to metabolic syndrome patients enhanced insulin sensitivity [41].

Glucagon-like peptide1 (GLP1) is associated with appetite and insulin secretion.Intestinal microbiota have been implicated in lowering levels of GLP1 and thereby slowing intestinal transit [42]. However, alterations of the microbiome [43] or bariatric surgry [44-46] decrease adiposity and increase GLP1 levels in mice.This is primarily attributed to butyrate production by commensal bacteria which can induce GLP1 production by intestinal luminal cells. [43].

Butyrate is propsed to increase the expression of the hormone angiopoietin-like protein4 (Angptk4), also known as fasting-induced adipose factor, a hormone implicated in the regulation of glucose and insulin sensitivity and lipid metabolism, inhibiting Lipoprotein Lipase (LPL) and thereby reducing fat storage. Despite the general trend toward repression of Angptl4 by the microbiota, specific bacteria can increase hormone expression. Mice treated with L. paracasei were leaner than controls, had lower ciculating lipids and elevated levels of Angptl4 [47]. This is probably mediated by butyrate. So, butyrate may play a role in the microbiota-induced weight maintenance changes that involve hormonal changes.

One interesting mechanism by which microbiota affect peptide hormones is through autoantibodies. Fetissov et al. [49] found that autoantibodies against peptide hormones involved in appetite control exists in healthy humans and rats, and affect feeding and anxiety. In GF rats, levels of these autoantibodies are altered, suggesting a novel mechanism by which the microbiome can affect appetite. These findings may have implications for the potential role of the microbiota in eating disorders such as anorexia nervosa and healthy controls.

New data among microbiota composition come from studies of gastric bypass surgery, in which the relative abundance of Gammaproteobacteria (Escherichia) and Verrucomicrobia (Akkermensia) is increased. While, microenvironment changes such as reduced food intake and reduction of bile acids, this is likely due to alterations in the levels of GIP (gastrointestinal inhibitory peptide), GLP1 and insulin following surgery [45,46,50-52].

Immune function

Gut microbiota play a role in modulating the immune response, both locally and systemically, beyond repressing pathogenic microbes [53]. In the absence of commensal bacteria, GF mice have impaired development of the innate and adaptive immune system [54-57]. reduced number of IgA producing plasma cells [58], and a decreased percentage of CD4+ T cells [59]. Additionally, T helper 17 Th 17 cells which produces proinflammatory cytokines are regulated by gut bacteria and are promoted specifically by segmented filamentous bacteria (SFB) [60]. Autoimmune Disease (AD) has been correlated with alterations of the microbiome (dysbiosis) The most extensively studied example is type 1 diabetes [61,62]. A different example linking the microbiota hormones and immunity comes from a study in mice, which showed that L. reuteri enhances wound -healing properties in the host through up-regulation of the neuropeptide hormone oxytocin, by a vagus nerve-mediated pathway [63].

Sepsis

Microbiome disruption may have a key role in sepsis and Acute Respiratory Distress Syndrome (ARDS). Dickson et al. have found culture-independent evidence that the lung microbiome is enriched with gut bacteria, both in a murine model of sepsis and in patients with ARDS (n=68). In more severely critically ill patients, lung bacteria were more outnumbered by the misplaced gut bacteria [64]. A large rural Indian trial (n=4556) showed the combination of the probiotic Lactobacillus plantarum plus the prebiotic fructooligosaccharide can help prevent sometimes deadly cases of sepsis and decrease lower respiratory tract infections in newborns. Panigrahi et al. [65] found that the synbiotic combination, which costst only one dollar per treatment, reduced neonatal sepsis and death by 40% from 9% in the placebo arm to 5, 4% among babies given the experimental treatment. This report underscores the importance of gut colonization on the maintenance of optimal immunological function.

It is believed intestinal microbiota not only act as a key defense system by locally supporting mucosal immunity, but also have proposed modulatory effects on systemic immunity. Schuyt et al. found that the gut microbiota play a protective role for the host during pneumococcal pneumonia, as reflected by increased bacterial dissimination, inflammation, organ damage and mortality in microbiota-depleted mice compared to controls. Fecal microbiota transplantation in gut microbiota-depleted mice restored local host defense. Whole genome mapping of alveolar macrophages showed up-regulation of metabolic pathways in the absence of a healthy gut microbiota. The up-regulation correlated with an altered cellular responsiveness, reflected by a reduced response to Lipopolysaccharide (LPS) and lipoteichoic acid. Compared to controls, alveolar macrophage derived from gut microbiota-depleted mice showed a diminished capacity to phagocytose S. pneumoniae [66].

The microbial ecosystems of the gut and the lungs change substantially in critically ill patients, resulting in dramatic changes to bacterial communities. In animal studies of shock the microbial contents of the gut determine the severity of multiorgan failure and the risk of death, an observation supported by trials of selective manupulations of the gut microbiome [67]. The mechanisms that drive gut-derived sepsis are incompletely understood and multifactorial, offering numerous unexplored therapeutic targets. During lung injury, the bacterial ecosystem of the alveolar shifts to a state of abundance in nutrients and growth-promoting host stress signals, leading to a positive feedback loop of inflammation and dysbiosis. The microbiome is a key therapeutic target for the prevention and treatment of critical illness [67]. However, large knowledge gaps remain [68].

Miscellaneous

Growth: No direct connection has been shown to date between the microbiota and growth hormones. The microbiome’s effect on grhelin and sex hormones may indirectly promote release of growth hormones [69]. Additionally, SCFA’s have been shown to inhibit growth hormones in cows, by affecting gene transcription in a cAMP/PKA /CREB- mediated signalling pathway [70]. Furthermore, bacteria produce somatostatin, which is a known growth inhibitor [71].

Sex hormones: Results regarding the relationship between sex hormones and the microbiota and vice versa are inconclusive. For example, Prevotella intermedius takes up estradiol and progesterone, which enhances its growth [72] Changes in expression of the estrogen receptor, ER-beta, also affect the intestinal microbiota composition [73]. This interaction goes both ways, as several types of bacteria have also been implicated in steroid secretion or modification [74]. For example Clostridium scindens converts glucocorticoids to androgens [74]. Intestinal bacteria also play a role in estrogen metabolism, because use of antibiotics leads to lower estrogen levels [75].

Pheromones: Pheromones are hormones that play important roles in sexual recognition, attraction and mating behavior as well as agression behavior and dominance. Pheromones are also termed ectohormones, chemicals secreted outside of the body of one individual and affecting the behaviors of others. In Drosophilia studies pheromones were affected by antibiotics and levels were related to a specific gut microbe [76]. These findings suggest a mechanism, whereby the microbiota affect host pheromone levels. Human date relating pheromones to the human microbiome are not available until now. Anyway, there are still considerable doubts about the existence of the human counterpart of putative pheromones. Actually, forty years of research of putative pheromones in humans is inconclusive and reserch in human pheromones should make a restart from scratch [77].

Treatment: Our perception of the microbiome has chamged rapidly the last decade, due to the metagenomic sequencing of the DNA and RNA repertoire present in the intestinal ecosystem and the re-emergence of gnotobiotic approaches enabling controlled microbial colonization of a mammalian intestine [78]. In contrast to the host’s genome, the microbial metagenome is highly dynamic and amenable to change over an individual’s lifetime [79]. Assuming a metagenomic contribution to disease susceptibility, this contribution is not stable, but rather undergoes fluctuations over time and depends on environmental inputs, that modulates its constitution [80]. Therefore, the therapeutic modulation of the microbiome might be harnassed to alter an individual’s risk for the manifestation of a certain disease. To design dietary or biotic interventions microbiota composition should be better understood [67,68].

One prototype microbiome-based intervention has recently been introduced in clinical practice as Fecal Microbiota Transplantation (FMT). Fecal microbiote transplantation is used in case of recurrent intestinal infection with antibiotic-resistant Clostridium difficile [81]. In the last 5 years FMT has become a widespread and broadly recommended approach in the treatment of recurrent C. difficile infections. Although standardization efforts are still underway, the procedure typically involves a certain level of donor screening [82]., sample homogenization and filtration, followed by administration via retention enema, endoscopy, nasogastric, or nasojejunal tubing, or in recently developed capsule formula. Several hundred cases of successful FMT have been reported , with cure rates up to 90% [83]. Despite the success and clinical effectiveness, the procedure remains poorly controlled. FMT involves the transfer of a large number of bacteria, viruses, and unicellular and multicellular eukaryotes, the individual function of which is largely unknown [84]. Such functions can manifest in phenotypic consequences, as seen in a case of unexpected weight gain, reported after familial FMT [84]. Also, in some cases, it might be the non-bacterial rather than the bacterial content, that mediates the efficacy of FMT. This has been exemplified by filtrated fecal transfer, in which only bacterial cell components, bacterial derived molecules, and viruses are retained [85]. Thus, more exact knowledge about interventions through specific microorganisms that mediate the benificial effects of FMT is crucial.

The success of FMT in treating recurrent pseudomembranous colitis has given rise to the hope that a similar procedure might prove effective against either intestinal or even extra-intestinal diseases. Indeed, cases of FMT trials have since been reported not only for gastrointestinal and infectious conditions, but also for metabolic, autoimmune, hematologic, and even neurologic conditions [86]. However, in contrast to recurrent C. difficile infections, the data from these trials are not sufficiently conclusive to recommend the immediate inclusion of FMT in standard clinical practice [87]. For instance, in the case of Inflammatory Bowel Disease (IBD), FMT has not yet proven to be the”magic bullet” in the form of a long awaited therapy across different manifestations of the disease, despite the fact that the microbiome is clearly involved in disease etiology. One reason could be that in IBD, the microbial community is not so disrupted as in recurrent pseudomembranous colitis after heavy prior antibiotic use. Another reason could be the microbiome in IBD is changing by environmental changes making it less amenable for FMT. Also, the microbiome in IBD might have enormous interindividual variations [88].

If FMT is not suitable for most microbiome-based therapeutic developments, what are the potential alternatives? One of the approaches could be the refinement of microbiotic engineering by more targeted approaches, selecting a single bacterium, that is as powerful as FMT-based community replacement, with respect to a clinically desired effect. Indeed in the case of C.difficile infection, this may be possible with only one strain, the already mentioned Clostridium scindens, which effectively inhibited C.difficile via the production of secondary bile acids in a rodent model [89].

Further developments of this strategy include the biological engineering of biotic interventions through system biology approaches in bacteria to enhance their functionallity [90]. Additonally, targeted interventions with the microbial ecosystem could be achieved through bacteriophages, a prominent component of the intestinal microbiome, with the capacity to re-gut the microbial gene pool [91]. Indeed, several clinical trials employing bacteriophage strategies are underway and have so far proven safe in the first phases [92].

However, the establishment of such viral therapies would necessitate an improved understanding of ecological interactions between the bacterial and bacteriophage communities in the intestine [93] and proof of efficacy [94].

As most modern drugs find their origin in endocrinology, a pharmacological approach could be based on future research in microbial endocrinology [95].

Conclusion

Microbial endocrinology shares the presence of neurochemicals that are exactly the same as in neurobiology of the host as well as in the microorganism. More clues to the existence of crosstalks between bacteria came from the discovery of the inter-kingdom, including the hormonal communication between microorganisms and their hosts. It appeared from this initial observation that bacteria perform quorum sensing. Endocrine effects of bacteria influence a variety of host responses, including behavior, metabolism and appetite and immune response. Butyrate may play a role in gastrointestinal hormone expression. Microbiota can also produce autoantibodies, increasing the expression of peptide hormones. New data on microbiota came from studies of gastric bypass surgery. Gut microbiota plays a role in modulating the immune response. Microbiome disruption may have a key role in sepsis and ARDS.

Our perception of the microbiome has changed rapidly the last decade, due to the metagenomic sequencing of the DNA and RNA repertoire present in the intestinal ecosystem and the re-emergence of gnotobiotic approaches. Fecal Microbiota Transplantations (FMTs) showed a cure rate up to 90% in recurrent antibiotic resistant Clostridium difficile infections. Results of FMTs for inflammatory bowel disease are less convincing. If FMT is not suitable for most microbiome-based therapeutic developments, refinement of microbiotic engineering by selecting a single bacterium could be a solution. Bacteriophage treatment is another possibility, but this needs an improved understanding of ecological interactions between bacterial and bacteriophage communities in the intestine. Also, bio-functionallity of bacteria can be enhanced.

As most modern drugs find their origin in endocrinology, a pharmacological approach could be based on future research in microbial endocrinology.

  1. Reiche EM, Nunes SO, Morimoto HK. Stress, depression, the immune system and cancer. Lancet Oncol. 2004; 5: 617-25. https://goo.gl/uqrLsp
  2. Lyte M. Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol 2004; 12: 14-20. https://goo.gl/FnyLk6
  3. Freestone P. Communication between bacteria and their hosts. Scientifica. 2013; 15. https://goo.gl/GhJny6
  4. Goldstein DS, Eisenhofer G, Kopin IJ. Sources and significance of plasma catechols and their metabolites in humans. J Pharm Exp Ther. 2003; 305: 800-11. https://goo.gl/cDfGhz
  5. Naafs Michael AB. Second Messengers in Endocrinology: A Mini-Review of the Cyclic Nucleotides. Endocrinol Metab Int J. 2017; 5: 00144. https://goo.gl/xunzzG
  6. Costa M, Brookes SJ, Hennig GW. Anatomy and physiology of the enteric nervous system. Gut. 2000; 47: 15-19. https://goo.gl/29kmCW
  7. Furness JB. The Enteric Nervous System. Malden, MA; USA: Blackwell Publishing; 2006. https://goo.gl/x7FaeH
  8. Lyte M. The role of microbial endocrinology in infectious disease. J. Endocrinol. 1993; 137: 343-5. https://goo.gl/172MBm
  9. Lyte M, Bailey MT. Neuroendocrine bacterial interactions in a neurotoxin-induced model of trauma. J Surg Res. 1997; 70: 195-201. https://goo.gl/2roaYV
  10. Iyer LM, Aravind L, Coon SL, Klein DC, Koonin EV. Evolution of cell-cell signalling in animals: did late horizontal gene transfer from bacteria have a role? Trends Genet. 2004; 20: 292-9. https://goo.gl/ABv5xj
  11. Hughes DT, Sperandio V. Inter-kingdom signalling: communication between bacteria and their hosts. Nat Rev Microbiol. 2008; 6: 111-20. https://goo.gl/wzy4qg
  12. Fuqua C, Winans SC, Greenberg EP. Census and consensus in bacterial ecoystems:the LuxR-Lux L family of quorum sensing transcriptional regulators. Annu Rev Microbiol. 1996; 50: 727-51. https://goo.gl/1KFPkg
  13. Karavolos MH, Winzer K, Williams P, Khan CM. Pathogene spionage: multiple bacterial adrenergic sensors eavesdrop on host communication systems. Mol Microbiol. 2013; 87: 455-65. https://goo.gl/AhYQ8U
  14. Sperandio V, Torres AG, Jarvis B, Nataro JP, Kaper JB. Bacteria-host communication: the language of hormones. Proc Natl Acad Sci U S A. 2003; 100: 8951-6. https://goo.gl/tL3Rdm
  15. erdoy M, Webster JP, Macdonald DW. Fatal attraction in rats with Toxoplasma gondii. Proc Biol Sci. 2000; 267: 1591-94. https://goo.gl/hpFKuK
  16. Blanchard EB, Scharff L, Schwarz SP, Suls JM, Barlow DH. The role of anxiety and depression in the irritable bowel syndrome. Behav Res Ther. 1990; 28: 401-05. https://goo.gl/NncDs3
  17. Wood JD. Enteric neuroimmunophysiology and pathophysiology. Gastroenterology. 2004; 127: 635-57. https://goo.gl/Jm8GLC
  18. Lyte M, Varcoe JJ, Bailey MT. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiol Behav. 1998; 65: 63-68. https://goo.gl/KMhtoC
  19. Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M. Activation in vagal afferents and central autonomic pathways: early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005; 19: 334-344. https://goo.gl/SuYppK
  20. Furness JB. The enteric nervous system and neurogastroenterology. Nat Rev Gastroenterol Hepatol. 2012; 9: 286-294. https://goo.gl/Z9fMTt
  21. Green BT, Lyte M, Chen C, Xie Y, Casey MA, Kulkarni Narla A, et al. Adrenergic modulation of Escherichia coli O157: H7 adherance to the colonic mucosa. AM J Physiol Gastroentest Liver Physiol. 2004; 287: 1238-1246. https://goo.gl/VJjnrA
  22. Breer H, Eberle J, Frick C, Haid D, Widmayer P. Gastrointestinal chemosensation: chemosensory cells in the alimentary tract. Histochem Cell Biol. 2012; 138: 13-24. https://goo.gl/MuoGSV
  23. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011; 23: 255-64. https://goo.gl/NRzJUb
  24. Yurdaydin C, Walsh TJ, Engler HD, Ha JH, Li Y, Jones EA, et al. Gut bacteria provide precursors of benzodiazepine receptor ligands in a rat model of hepatic encephalopathy. Brain Res. 1995; 679: 42-48. https://goo.gl/F2LThR
  25. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulators emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011; 108: 16050-16055. https://goo.gl/nQHvkZ
  26. Li W, Dowd SE, Scurlock B, Acosta-Martinez V, Lyte M. Memory and learning behavior in mice is temporally associated with diet-induced alterations in gut bacteria. Physiol Behav. 2009; 96: 557-567. https://goo.gl/juecgC
  27. Douglas Escobar M, Elliott E, Neu J. Effect of intestinal microbial ecology on the developing brain. JAMA Pediatr. 2013; 167: 374-379. https://goo.gl/HnAkCA
  28. Cho I, Yamanishi S, Cox L, Methé BA, Zavadil J, Li K, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012; 488: 621-626. https://goo.gl/NJ1UpS
  29. Cherbut C, Ferrier L, Roze C, et al. Short -chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am J Physiol. 1998; 275: 1415-1422.
  30. Fukumoto S, Tatewaki M, Yamada T, Fujimiya M, Mantyh C, Voss M, et al. Short-chain fatty acids stimulate coloinic transit via intraluminal 5-HT release in rats. Am J Physiol Regul Integr Comp Physiol. 2003; 284: 269-276. https://goo.gl/rU8SEv
  31. LeRoith D, Pickens W, Vinek, et al. Bacillus subtilus contains multiple forms of somatostatin -like material. Biochem Bioph Res Co. 1985; 127: 713-719.
  32. Lam V, Su J, Koprowski S, Hsu A, Tweddell JS, Rafiee P, et al. Intestinal microbiota determine severity of myocardial infarction in rats. FASEB J. 2012; 26: 1727-1735. https://goo.gl/CU3382
  33. Ravussin Y, Koren O, Spor A, LeDuc C, Gutman R, Stombaugh J, et al. Responses of gut microbiota to diet composition and weight loss in lean and obese mice. Obesity. 2012; 207: 738-747. https://goo.gl/DeKLyG
  34. Queipo Ortuno MI, Seoane LM, Murri M, Pardo M, Gomez Zumaquero JM, Cardona F, et al. Gut microbiota composition in male rat models under different nutritional status and physical activity and the associatiion with serum leptin and grhelin levels. PloS One 2013; 8: 654-665. https://goo.gl/ZFssxw
  35. Rajala MW, Patterson CM, Opp JS, Foltin SK, Young VB, Myers MG Jr. Leptin acts independently of food intake to modulate gut microbial composition in male mice. Endocrinology. 2014; 155: 748-75. https://goo.gl/TwrL7M
  36. Takamura N, Okubo T, Sonoyama K. Lactobacillus plantarum strain no 14 reduces adipocyte size in mice fed high-fat diet. Exp Biol Med. 2010; 235: 849-856. https://goo.gl/zAWySR
  37. Naruszewicz M, Johansson ML, Zapolska-Downar D, Bukowska H. Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers. Am J Nutr. 2002; 76: 1249-1255. https://goo.gl/1UAViQ
  38. Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased grhelin and increased peptide YY in overweight and obese adults. Am J Clin Nutr. 2009; 89: 1751-1759. https://goo.gl/QrS9A6
  39. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012; 490: 55-60. https://goo.gl/BEyZNa
  40. Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013; 498: 99-103. https://goo.gl/qRLERi
  41. Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JF, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012; 143: 913-916. https://goo.gl/4qqipy
  42. Wichmann A, Allahyar A, Greiner TU, Plovier H, Lundén GÖ, Larsson T, et al. Microbial modulation of energy availability in the colon regulates intestinal transit. Cell Host Microbe. 2013; 14: 582-590. https://goo.gl/beCQ6K
  43. Yadav H, Lee JH, Lloyd J, Walter P, Rane SG. Benificial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. J Biol Chem. 2013; 288: 88-97. https://goo.gl/H6ipBg
  44. Zhang H, DiBaise JK, Zuccola A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci. 2009; 106: 65-70. https://goo.gl/3V1iNQ
  45. Liou AP, Paziuk M, Luevano JM, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013; 5: 78-141. https://goo.gl/Aksxbk
  46. Osto M, Abegg K, Bueter M, le Roux CW, Cani PD, Lutz TA. Roux and Y gastric bypass surgery in rats alters gut microbiota profile along the intestine. Physiol Behav. 2013; 119: 92-96. https://goo.gl/RBWem9
  47. Aronsson L, Huang Y, Parini P, Marion Korach-André, Janet Håkansson, Jan-Åke Gustafsson, et al. Decreased fat storage by Lactobacillus paracasei is associated with increased levels of angiopoietin-like 4 protein (ANGPTL4). Plos One. 2010; 5: 13087. https://goo.gl/G1trjz
  48. Fettisov SO, Hanze Sinno M, Coeffier M, Bole-Feysot C, Ducrotté P, Hökfelt T, et al. Autoantibodes against appetite-regulating peptide hormones and neuropeptides: putative modulation by gut microflora. Nutrition.  2008; 24: 348-359. https://goo.gl/jrNriL
  49. Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. Plos One.  2009; 7125. https://goo.gl/FpA9N9
  50. Laferre B. Do we really know why diabetes remits after gastric bypass surgery? Endocrine. 2011; 40: 16-27. https://goo.gl/JuYa3F
  51. Madsbad S, Dirksen C, Holst JJ. Mechanisms of changes in glucose metabolism and bodyweight after bariatric surgery. Lancet Diabetes Endocrinol. 2014; 2: 152-164. https://goo.gl/jdd2J2
  52. Wen L, Duffy A. Factors Influencing the Gut Microbiota, Inflammation and Type 2 Diabetes. J Nutr. 2017; 147: 1468-1475. https://goo.gl/eQUys1
  53. Kamada N, Seo SU, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013; 13: 321-335. https://goo.gl/jA3Rir
  54. Hill DA, Artis D. Intestinal bacteria and the regulation of immune cell homeostasis. Annu Rev Immunol. 2010; 28: 623-667. https://goo.gl/Y4voYt
  55. Littman DR, Palmer EG. Role of the commensal microbiota in normal and pathogenic host immune response. Cell Host Microb. 2011; 10: 311-323. https://goo.gl/pe9H7b
  56. Honda K, Littman DR. The micobiome in infectious disease and inflammation. Ann Rev Immunol. 2012; 30: 759-795. https://goo.gl/DZyMEQ
  57. Hooper LV, Littman DR, MacPherson AJ. Interactions between the microbiota and the immune system. Science. 2012; 336: 1268-1273. https://goo.gl/qQyiaM
  58. Crabbe PA, Nash DR, Bazin H, Eyssen DV, Heremans JF. Antibodies of the IgA type in intestinal plasma cells of germ-free mice after oral or parenteral immunization with ferritin. J Exp Med. 1969; 130: 732-744. https://goo.gl/gBD17c
  59. Ostman S, Rask C, Wold AE, Hultkrantz S, Telemo E. Impaired regulatory T cell function in germ-free mice. Eur J Immunol. 2006; 36: 2336-2346. https://goo.gl/HMogJL
  60. Tanabe S. The effect of probiotics and gut microbiota on Th 17 cells. Int Rev Immunol. 2013; 32: 511-525. https://goo.gl/QcJjHg
  61. Brown CT, Davis-Richardson AG, Grongo A, Gano KA, Crabb DB, Mukherjee N, et al. Gut microbiome metagenomics analysis suggests a functional model to the development of autoimmunity for type 1 diabetes. Plos One. 2011; 6: 625-792. https://goo.gl/bNxfW9
  62. Hare N, Alkamani AK, Ir D, Robertson CE, Wagner BD, Frank DN, et al. The role of the intestinal microbiota in type 1 diabetes. Clin. Immunol. 2013; 146: 11-29. https://goo.gl/UEm3k2
  63. Pontahidis T, Kearney SM, Levkovich T, Peimin Qi, Bernard J Varian, Jessica R Lakritz, et al. Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. Plos One. 2013; 8: 78898. https://goo.gl/YKpRGH
  64. Dickson RP, Singer BH, Newslead MN, Nicole RF, John RE, Theodore JS, et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nature Microbiology. 2016; 113. https://goo.gl/18zs2r
  65. Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017; 548: 407-412. https://goo.gl/8fW2jm
  66. Schuyt TJ, Lankelma JM, Brendon P, de Sousa e Melo F, Roelofs JJ, de Boer JD, et al. The gut microbiota plays a protective role in the host defense against pneumococcal pneumonia. Gut. 2016; 65: 575-583. https://goo.gl/SMzLwu
  67. Dickson RP. The microbiome and critical illness. Lancet Respir Med. 2016; 4: 59-72. https://goo.gl/UpeDXA
  68. Haak BW, Wiersenga J. The role of the gut microbiota in sepsis. Lancet Gastroenterology & Hepatology.  2017; 2: 135-143. https://goo.gl/65XtNz
  69. Howard AD, Freighner SD, CullyD, Arena JP, Liberator PA, Rosenblum CI, et al. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science. 1996; 2: 739-747. https://goo.gl/YZUVLc
  70. Wang JF, Fu SP, Li S, Hu ZM, Xue WJ, Li ZQ, et al. Short-chain fatty acids inhibit growth hormone and prolactin gene transcription via Camp/PKA/CREB signalling pathway in dairy cow anterior pituitary cells. Int J MolSci. 2013; 14: 474-488. https://goo.gl/8b93ZB
  71. LeRoith D, Pickens W, Vinik AI. Shiloach J. Bacillus subtilus contains multiple forms of somatostatin-like material. Biochem Biophys Res Co. 1985; 121: 713-719. https://goo.gl/9wQecv
  72. Kornman KS, Loesche WJ. Effects of estradiol and progesterone on Bacteroides metaninogenicus and Bacteroides gingivalis. Infect Immun. 1982; 35: 256-263. https://goo.gl/CD4Sdx
  73. Menon R, Watson SE, Thomas LN, Allred CD, Dabney A, Azcarate-Peril MA, et al. Diet complexiity and estrogen receptor beta status affect the composition of the murine intestinal microbiota. Appl Environ Microb. 2013; 79: 63-73. https://goo.gl/mXAmBU
  74. Ridlon JM, Ikegawa S, Alves JM, Zhou B, Kobayashi A, Iida T, et al. Clostridium scindens: a human gut microbe with a high potential to convert glucocorticoids into androgens. J Lipd Res. 2013; 54: 37-49. https://goo.gl/RZtESK
  75. Adlercreutz H, Pulkinnen MO, Hamalainen EK, Korpela JT. Studies on the role of intestinal bacteria in metabolism of synthetic and natural steroid hormones. J Steroid Biochem. 1984; 20: 217-229. https://goo.gl/He48uT
  76. Sharon G, Segal D, Ringe JM, Hefetz A, Zilber-Rosenberg I, Rosenberg E. Commensal bacteria play a role in mating preference of Drosiphila melanogaster. Proc Natl Acad Sci. 2010;107: 51-56. https://goo.gl/j11rHK
  77. Naafs Michael AB. Pheromones: Honey’s and Queens. Glob J Otol. 2017; 9. https://goo.gl/ajMiQn
  78. Tambaugh PJ, Gordon JL. An invitation to the marriage of metagenomics and metabolomics. Cell. 2008;134: 708-713. https://goo.gl/LDfQjc
  79. David LA, Materna AE, Friedman J, Maria I Campos-Baptista, Matthew CB, Allison P, et al. Host life-style affects human microbiota on daily timescales Genome. Biol. 2014; 15: 89. https://goo.gl/cFmkWp
  80. Khosravi A, Yunez A, Price JG, Chow A, Merad M, Goodridge HS, et al. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe. 2014;15: 374-381. https://goo.gl/fkP3Zo
  81. van Nood E, Vrieze A, Nieuwdorp M, Susana Fuentes, Erwin GZ, Willem M  de Vos, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013; 368: 474-475. https://goo.gl/L8E3zR
  82. Moayyed P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, et al. Fecal microbiota transpantation induces remission in patients with active Ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015; 149: 102-109. https://goo.gl/ihUn36
  83. Newman KM, Rank KM, Vaughn BP, Khoruts A. Treatment of recurrent C. difficile infection using fecal microbiota transplantation in patients with inflammatory bowel disease. Gut Microbes. 2017; 8: 303-309. https://goo.gl/W9ju94
  84. Alang N, Kelly CR. Weight gain after fecal microbiota transplantation. Open forum infectious diseases. 2015. https://goo.gl/98yR7A
  85. Ott SJ, Wactzig GH, Rehman A, Moltzau AJ, Bharti R, Grasis JA, et al. Efficacy of sterile filtrate transfer for treating patients with Clostridium difficile infection. Gastroenterology. 2016; 152: 799-811. https://goo.gl/UPNBtC
  86. Cohen NA, Manarshak N. Novel identification for fecal transplantation: update and review of the literature. Dig Dis Sci. 2017; 1131-1145. https://goo.gl/fprrmR
  87. Reinisch W. Fecal microbiota transplantation in inflammatory bowel disease.  Dig Dis. 2017; 35: 123-126. https://goo.gl/yjmCX5
  88. Luzepone CA, Stombaugh JL, Gordon JL, Jansson JK, Knight R. Diversity, stability and resilence of the human gut microbiota. Nature. 2012; 489: 220-230. https://goo.gl/aZoLbd
  89. 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: 205-208. https://goo.gl/5sJyX7
  90. Mimee M, Tucker AC, Voigt CA, Lu TK. Programming a human commensal bacterium, Bacteroides thetaiotamicron, to sense and respond to stimuli in the murine gut microbiota. Cell Syst. 2015; 1: 62-71. https://goo.gl/A3LhMY
  91. Brussow H. Biome engineering-2020. Microb Biotechnol. 2016; 9: 553-563. https://goo.gl/VcXq3R
  92. Vanden HD, Lavigne R, Brussow H. Bacteriophage therapy: advances in formulation strategies and human clinical trials. Annu Rev Virol. 2015; 2: 599-618. https://goo.gl/wjmHgT
  93. Knowles B, Silveira CB, Bailey BA, Barott K, Cantu VA, Cobián Güemes AG, et al. Lytic to temperate switching of viral communities. Nature. 2016; 531: 466-470. https://goo.gl/3rQtq5
  94. Sarker SA, Sultana S, Renteler G, Moine D, Descombes P, Charton F, et al. Oral phage therapy of acute diarrhea with two coliphage preparations: a randomized trial in children from Bangladesh. Ebiomedicine. 2016; 4: 124-137. https://goo.gl/F8VJz1
  95. Naafs, Michael AB. Pharmacodynamic Evaluation: Endocrinology; Chapter 35. In Drug Discovery and Evaluation: Methods in Clinical Pharmacology, 2nd Edition, 2017. Editors: Hock FJ, Gralinski MR. Springer Verlag, Berlin, Heidelberg, New York.