Open access

Glucocorticoids and the Intestinal Environment

Written By

Hümeyra Ünsal and Muharrem Balkaya

Submitted: 23 April 2012 Published: 28 November 2012

DOI: 10.5772/51977

From the Edited Volume

Glucocorticoids - New Recognition of Our Familiar Friend

Edited by Xiaoxiao Qian

Chapter metrics overview

1,966 Chapter Downloads

View Full Metrics

1. Introduction

For thousands, perhaps millions of years, human have marveled to understand how the body is formed and function, how it maintains its wholeness and healthy state, and how disordered states occur. In frame of holistic philosophy of ancient eastern cultures, a person had been accepted as physical, emotional, mental, and spiritual ones. In this concept, the balance of the body, mind and spirit were accepted as the healthy state of an individual [1,2]. The ancient Greeks defended the concept of the four humors doctrine; blood, black bile, yellow bile, and phlegm. Ad modum this doctrine these four humors in a healthy person were believed to be balanced, and the reason why a person fell ill was due to the fact that these fluids were disturbed [3,4]. With the separation of medicine from superstition, magic and religion by famous Greek physician Hippocrates in around 5th century B.C., more realistic and relevant approaches could be developed to better understanding the life, structures of living creatures and their healthy and diseased conditions. These and the time-dependent innovative developments in technology and science throughout the following centuries led to more concrete observations, mainly on animals. Gathered evidences and additive knowledge base from these observations mankind led mankind to make realistic definitions on the subjects “the structure, integrity and functions” of the animal and human body. In 19th century Louis Pasteur, a French chemist and microbiologist, stated the germ theory of diseases. He believed that micro-organisms (bacteria) infect animals and humans, thus they cause diseases [5]. At the same times, a French Physiologist Claude Bernard tried to understand how living creatures maintain their integrity, and how it is regulated, and defeated. He discriminated the internal environment from the external environment and was in opinion that living creatures maintain their internal milieu relatively constant under continuously changing environmental conditions. Claude Bernard [6] attributed also an important role to nervous system in the maintenance of internal environment in physiological ranges in human and animal organisms and remarked the substantial differences between animals and plants in this respect. Since that time, the interest of the researchers in different fields worldwide is mainly focused on possible regulation mechanisms of various living species. Thus, it can be said that the philosophical roots of the concept “stress” and “stress physiology” is going back to the early observations that living creatures are exposed continuously to the effects of various environmental challenges against which they have to defend their integrity, and following statement from Claude Bernard that living creatures strive to maintain their internal environments relatively constant via various homeokinetic mechanisms even if their environmental conditions are changing, thereby keep their normal physiological functioning and prerequisite for a free, independent live or shortly ‘la fixité du miliéu interieur est la condition d'une vie libre et indépendante’ as defined by him “Il y a pour l'animal deux milieux: un milieu extérieur dans lequel est placé l'organisme, et un milieu intérieur dans lequel vivent les éléments des tissus”. However, Hans Selye, one of the Pioneers and founders of stress from 1930’s, was the first who introduced the term “stress” as the real or perceived physical or psychological events which are threaten the homeokinesis in medical terminology. Ad modum Selye the stress is somewhat like living, not so easy to define, although there is no doubt about its presence [7]. The environmental challenges affecting a living creature, thus causing stress, can vary from physical, to chemical and bio-psycho-social factors, while all the reactants, their contra- and/or co-players within the living systems are of chemical nature at the last instance; intracellular or intra-bodily signaling elements as either hormones including glucocorticoids and catecholamines or neurotransmitters, cytokines and chemokines, etc., or a group or all of them functioning simultaneously within the body for the same purposes.

Stress and the glucocorticoids are associated or interweaving concepts with each other. Indeed, to physiologists the term “stress” has come to mean any event that elicits increased cortisol secretion [8]. However, as it is well known, glucocorticoids are not only mediators of the stress responses; they take a part in peripheral components of stress responses. The stress response is mediated by the stress system which is composed of two components; central nervous system and the peripheral part. The central, greatly interconnected effectors of this system include the hypothalamic hormones arginine vasopressin, corticotropin-releasing hormone/factor (CRH/CRF) and pro-opiomelanocortin-derived peptides, and the locus ceruleus and autonomic norepinephrine centers in the brainstem. The peripheral components of the stress system include (a) the peripheral limbs of the hypothalamic-pituitary-adrenal (HPA) axis; (b) the efferent sympathetic-adrenomedullary system, and (c) components of the parasympathetic system [9-11]. There are also bidirectional positive feedback regulation between the CRF secreted paraventricular nucleus of hypothalamus and central noradrenergic system [9,11,12].

Stressors activate different physiological processes. The first classical response is the secretion of adrenalin and noradrenalin hormones from the adrenal medulla via activation of sympathetic nervous system. This response is called “fight or flight syndrome”, because adrenaline and noradrenalin increase the respiratory rate, the heartbeat, the concentration of glucose in circulating blood and the blood flow to the skeletal muscles. This fast response is primarily related to survival. Stressors also activate the HPA axis. The activation of this axis begins with the stimulation of parvocelular neurons of hypothalamus and secretion of CRH. CRH stimulates the secretion of adrenocorticotrophic hormone (ACTH) from the adenohypophsis and the ACTH acts on cortex of adrenal glands and stimulates the release of glucocorticoid hormones. GC hormones (corticosterone in rodents and cortisol in humans), which are the ultimate product of HPA axis activation, act on multiple bodily systems to maintain homeokinesis. They stimulate protein catabolism, gluconeogenesis and release of glycerol and fatty acids into the blood, maintain the vasoconstructive effect of norepinephrine, inhibit glucose uptake and oxidation by many body cells except the brain (insulin antagonism) and also inhibit inflammation and specific immune response [8-11].

The secretion of ACTH, and therefore of cortisol or corticosterone, is stimulated by several hormones and molecules in addition to hypothalamic CRH. Depending on the stressor, substances such as vasopressin, epinephrine, angiotensin II, various cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6), and lipid mediators of inflammation affect the hypothalamic, pituitary, and/or adrenal components of the HPA axis and potentiate its activity. Glucocorticoids play an important role in the regulation of basal activity of the HPA axis, as well as in the termination of the stress response by acting at extra-hypothalamic centers, the hypothalamus, and the pituitary gland. The negative feedback of glucocorticoids on the secretion of CRH and ACTH serves to limit the duration of the total tissue exposure of the organism to glucocorticoids, thus minimizing the catabolic, lipogenic, anti-reproductive, and immunosuppressive effects of these hormones [8,10,11].

The activation of the HPA is a longer-term adjustment by the humans or animals to the changes in their micro- or surrounding environments. Selye [7] called it as the ‘general adaptation syndrome’ (GAS). The process of adaptation, also known as “allostasis” (literally “maintaining stability, through change”), supports the homeokinesis [13]. In acute stress, the activation of both sympathetic system and HPA axis are essential for survival of individual and they help to re-establish or maintain homeokinesis through adaptation. However, in chronic periods, prolonged or repeated activation of stress systems can result in cumulative biological changes (known as allostatic load) and can alter adaptive mechanism. If the ability of the organisms to maintain their integrity is poor or not strong enough to compensate the effects of environmental challenges, they can also harm the living creatures via various mechanisms. Excessive or inappropriate, inadequate adaptive responses to stress may play a causal role in development of certain diseases [10,11,14,15]. Indeed, depending on the stressors and their severity as well as the capability of living creatures to respond them, the reactions of living creatures to the stressors can be systematic, also reflecting itself throughout the whole body, or mainly local or regional, reflecting self at the organel/celluler/organ or system levels [11,14]. Similarly, the application of glucocorticoids for a long time can also cause events with serious consequences [16].

The gastrointestinal tract and the immune system are particularly responsive to different stressors. This system has many cellular targets for the stress mediators such as catecholamines, glucocorticoids and CRF [17-19]. The association between stress and various gastrointestinal diseases, including functional bowel disorders, inflammatory bowel disease (IBD), peptic ulcer disease and gastroeosophageal reflux disease, is being actively investigated [15,17]. However, there is a parodox that the chronic stress plays a role in inflammatory bowel disease while the glucocorticoid therapy is widely used to cure the same disease.

Gastrointestinal system (GIS) is the biggest surface area (~200 m2) of the body that is binding the organism to the external environment. This system is continuously exposed to various antigens from consumed foods and resident bacteria and also from potentially harmful pathogens, such as viruses, bacteria, fungi or parasites. Besides digestion and absorption processes, it forms a barrier between the internal and external environments [20]. Intestinal barrier is composed of various components such as tight junction conformation between the intestinal epithelial cells, mucosal immune system, mucin secretion, and intestinal microbiota. The barrier function is quite critical process because of the barrier should confirm the hyporesponsiveness or tolerance towards commensal bacteria while maintaining the ability to fight pathogenic microorganisms. The breakdown of this critical balance usually results in inflammation-associated damages. Gastrointestinal system is rather complex system, which has own nervous and endocrine system. The enteric nervous system is connected bidirectionally to the brain by parasympathetic and sympathetic pathways forming the brain–gut axis. The description of stress-induced alterations in this axis is thought to be important for the solving problems of many stress-related gastrointestinal disorders [17,20]. A number of paracine acting endocrine cells have important functions for the regulation of digestion processes. These hormones exist in both central nervous system and gastrointestinal plexus neurons where they function as neurotransmitters or neuromodulators [8]. Recent findings suggest that glucocorticoid hormones may also be synthesized from extra-adrenal tissues such as brain, skin, vascular endothelium and intestine. These tissues express steroidogenic enzymes and were claimed to be potential extra-adrenal sources of glucocorticoids [21]. It is emphasized that intestinal glucocorticoid synthesis might be of potential importance in regulation of mucosal immune responses and information of inflammatory bowel disease [22].

Gastrointestinal system hosts also a large number of microorganisms. They all together are called as gastrointestinal microbiota, and form the great part of the system. The microbiota–host interactions play an active role in many physiologic and pathophysiologic processes of the host [19,23,24]. While gastrointestinal microbiota could affect the stress response [25] and intestinal structure and functions of the host [23,24], stress hormones of the host organism such as the catecholamines can also alter directly growth, motility, biofilm formation and/or virulence of pathogens and commensal bacteria [19,26,27] or they may affect their microbiota indirectly by changing the intestinal environment [20].

The effects of stress on important physiological functions of the gut include; motility, secretion and absorption, visceral sensitivity, mucosal integrity, permeability, immune system, blood flow, microbiota and microbiota-host interactions [17,20]. Some stress-related alterations in gut physiology also can be induced by exogenous glucocorticoids [28-30]. However; recent findings using central and peripheral CRF administration showed that CRF and their receptor subtypes (CRF1 and CRF2) are playing important roles in many stress-related alterations of the gut. The stress-related alterations including intestinal permeability, mast cell activity [20], goblet cell and mucin formation [31], and motility alterations [32] can be mimicked by the CRF agonists and inhibited by CRF receptor antagonists. Therefore; CRF have been suggested as a target to treat stress-induced functional gastrointestinal disorders.

In this chapter, the effects of stress and stress-related hormones, especially glucocorticoids on the components of intestinal environment suc as intestinal epithelium, mucin formation, mucosal immune system, intestinal microbiota and microbiota-host interactions, and also the role of corticosteroids and stress in bacterial colonization and intestinal diseases were reviewed.

Advertisement

2. Stress models for gastrointestinal studies

A great number of animal models of stress (acute/chronic, early life/adult, physical/ psychological or both physical and psychological) have been described for studying the effects of stress on gastrointestinal functions. For this purpose, rather naturally (Wistar-Kyoto rats) or genetically modified stress susceptible animals were also used. As denoted by Soderholm and Perdue [20], stress models that psychological effects are more powerful than the physical effects are preferred for mimicking the effects of the life and environmental stress on several pathologies. However, the kind and duration of stressors and other factors including genetic or perinatal environment, etc. might be a key factor in the evaluation of the stress effects on specific gastrointestinal functions such as stress-induced visceral hyperalgesia. Depending on the characteristics of the stressors and the time-course of their effects, alterations caused by stress can be immediate, delayed, transient or sustained or never be seen [33,34]. For example, the small intestinal transit was significantly inhibited by restraint stress, but not by footshock stress although plasma corticosterone levels were significantly elevated to the same extent by restraint stress and foot-shock stress [33].

Williams et al [35] reported that acute mild restraint (wrap restraint) elevated plasma levels of adrenocorticotropic hormone and beta-endorphin, and caused analgesia. Gastric ulcer did not form, gastric emptying was not affected, however, small intestinal transit was inhibited, and large intestinal transit was stimulated by wrap restraint stress, and there was an associated increase in fecal excretion. Because of neither adrenalectomy nor hypophysectomy have prevented the response of the intestine to stress, they suggested that adrenal or pituitary-derived factors are not responsible for mediating the effects of stress on the gut.

Restraint stress is most widely used as an acute stress model. Restraining can be supplied with restraint devices or by wrap restraint for times varying from 30 min to 4 hours. This model could be modified with a cold environment (cold restraint stress) or water immersion (water immersion restraint stress) for creating both physical and psychological stress [20]. Cold enhanced the changes in rat intestine caused by restraining. However, plasma corticosterone levels increased in both restraint and cold restraint stressed rats in same extent compared with fasted unstressed rats. Fasting corticosterone levels also increased compared with the fed rats [36].

The acute (one time) and chronic (1 hour/day for 10 consecutive days) models of water avoidance stress are also preferred potent psychological models in determination of stress-induced gastrointestinal functions. These models result in elevations of ACTH and corticosterone within 30 min [37], and induce enlargement of the adrenal glands [38]. Recently, Vicario et al [39,40] reported that crowding stress (8 rats/cage) or sham-crowding (2 rats/cage) for up to 15 consecutive days triggers reversible inflammation, mast cell-mediated barrier dysfunctions, persistent epithelial dysfunction, and colonic hyperalgesia. Crowding-stressed rats showed higher plasma corticosterone levels than sham-stressed animals from day 1 and up to day 15. After 15 days of crowding stress, corticosterone levels decreased %38 (slightly adaptation), but HPA reactivity to incoming stressors was preserved. Crowding stress, differently from the stress models induced in laboratory, is actualized in natural environment of animals and may reflect life stress well for humans. Thus, this model has been also suggested as a suitable animal model to unravel the complex pathophysiology underlying to common human intestinal stress-related disorders, such as IBS.

Early life stress models such as maternal deprivation of pups from the dams have also been widely used [31,41,43,44], because stressful events in the early period of life (in the form of abuse, neglect or loss of the primary caregiver) have been shown to modify adult immune and gastrointestinal tract functions [12].

Advertisement

3. Intestine and its microenvironment

The gastrointestinal system is a tubular organ and the part ‘intestine’ extends downwards from the pyloric sphincter. With its many loops and coils, small and large intestines fill much of the abdominal cavity. Microscopically, both the small and large intestine are composed of four distinct layers; the mucosa covering the internal site of the tube, the submucosa, the muscularis externa and the serosa. The mucosa consists of layer of epithelia, loose connective tissue layer (lamina propia) containing blood vessels, lymphatics, and some lymphoid tissue, and muscularis mucosa. They both have different types of cells as building blocks; however, some of them are especially important in relation to luminal challenges via different agents including undigested or partly digested dietary proteins, gut microbiota and its metabolites. Located among the enterocytes are goblet cells secreting mucin which per se build a barrier; scattered enteroendocrine cells with paracrine and endocrine actions; Paneth cells characterized by their granules containing lysozyme, tumor necrosis factor-α and defensins have antibacterial roles; M cells with their well-known roles in antigen sampling and transportation; and the intestinal subepithelial myofibroblasts located in proximity to the mucosal epithelium and produce growth factors including hepatocyte growth factor promoting the proliferation of the intestinal epithelial stem cells, thus responsible for regeneration and maintaining of the integrity of the intestinal mucosa; and dendritic cells (DCs) highly specialized for antigen presentation which can capture non-self proteins and recognize microbial products [45-55].

Intestinal epithelial cells continuously contact with two different environments; first, luminal environment, which include intestinal secretions, food antigens, commensal bacteria, and also noxious or pathogenic materials, and second, the interstitial fluid surrounding the cells at the basolateral side. The single layer intestinal epithelium along the small and large intestine has a number of physiologic functions; it forms a barrier between the external (luminal) and internal environments besides its digestive and absorptive functions, and this epithelial barrier limits the space for bacterial growth. The ability of the epithelium to control uptake of molecules into the body is denoted as the intestinal barrier function [20,56,57]. The characteristics of intestinal epithelium for participation to the barrier function include; tight junction adherence between the epithelial cells, fluid and mucin secretions, secretions of numerous antimicrobial peptides, transepithelial transport of secretory IgA and the antigen presenting cell activity. However, intestinal epithelium is not only component of the intestinal barrier. Mucosal immune system, microbiota and microbiota-host interactions exist in major components of intestinal barrier [23,24]. All these components of intestinal barrier are controlled by the mediators of neuro-endocrine-immune network, and stress and stress mediators have significant impact on these components and regulatory network [20,24].

Advertisement

4. The effects of stress and glucocorticoids on intestinal barrier function

Intestinal mucosal epithelium is very sensitive against different types of stress because of the half-time of mucosal epithelia which may be as short as one and half day in certain parts. In other words, compared to many other tissues in the human and animal body, it is not well differentiated and very fragile [8,57]. The effects of acute or chronic stress on intestinal barrier function or intestinal permeability does not appear very different from each other in animal models of stress. However, the duration and repetition of stressors may influence severity, and the alterations may be temporary or permanent [34,58]. Generally, intestinal ion secretion [40,59], macromolecular permeability [60,61], inflammation [40,59], visceral hypersensivity and colonic motility increased [40], while gastric motility decreased in various animal models of stress [62-64]. These stress responses have been also described in IBD patients and involve dysregulation of HPA axis [12,15,17]. They are mediated by stress-related neuropeptides such as CRF, neural mechanisms and mast cells [18,20,65]. Various stress factors including heat, nutritional alterations, overcrowding, physical restraints and transporting also destroy the microbial balance and their microenvironment in the gut [66,67]. When the stressful events cause a decrease in beneficial bacteria, they generally increase the pathogenic species within the gut microenvironment [67-69]. In following sections, the effects of stress mediators on each component of barrier were discussed in details.

Advertisement

5. The physical barrier of epithelium

Cell-cell and cell–basement membrane interactions of intestinal epithelial cells control the transcellular and paracellular transports of luminal macromolecules and prevent bacteria from translocating into the subepithelial layer. Tight junctions (TJ) are primary physical components of intestinal barrier, located at the most apical part of lateral membranes of epithelial cells and restrict paracellular passage. The breakdown of tight junctions during bacterial infections results in gut barrier failure, often termed “leaky gut” [20,23]. Proteins that constitute the TJ complex include transmembrane proteins such as occludin, claudin, junction adhesion molecules and intracytoplasmic proteins zonula occludens 1 and 2 (ZO1-ZO2) and members of the membrane-associated guanylate kinase (MAGUK) protein family [70]. Tight junctions are highly dynamic structures, and their permeability is regulated by several physiological and pathophysiological conditions. Signals from intestinal microbiota may promote integrity of the epithelial barrier and have been shown to regulate tight junctions and protect intestinal epithelial cells (IECs) from injury by controlling the rate of IEC proliferation and inducing cytoprotective proteins [23]. Inflammatory cytokines can disrupt tight junctions and impair gut barrier integrity. Treating epithelial monolayers with TNF-α or IL-1b increased the permeability of tight junctions by stimulating transcription and activation of myosin light chain kinase (MLCK) [71-73]. The acute partial wrap restraint stress increased colonic permeability and rectal hypersensitivity via epithelial cell cytoskeleton contraction through myosin light chain kinase activation [74]. Acute immobilization stress also induced an increase in TJ permeability in the rat terminal ileum. These changes were mainly due to irregularly distribution of TJ transmembrane protein occludin and of the plaque protein ZO-1 which were seen after 2 hours from the stress induction and returned to a normal pattern within 24 hours [70]. Mazzon and Cuzzocrea [75] also suggested that TNF-α has active roles in the increase of tight junction permeability during acute restraint stress. They demonstrated in vivo in a TNF-α R1 knock-out mouse (TNF-α R1KO) model of restraint stress that the inhibition of TNF-α attenuates the development of TJ alteration in the ileum. Restraint stress caused the increase of heat shock protein-70 expression and associated decrease in the expression of type 1 (ZO-1) protein in the colonic epithelium of mice. These stress-induced changes can be inhibited by the glucocorticoid receptor antagonist mifepristone [76]. However, Boivin et al. [77] reported that glucocorticoids enhanced epithelial barrier function by suppressing transcription of myosin light chain kinase. Bacterial pathogens target tight junctions and breach epithelial integrity to promote colonization, obtain nutrients and access the underlying tissues [23]. So, alterations in gastrointestinal microbiota induced by several stressors or exogenous glucocorticoids might be an important threat for the barrier disruption.

Advertisement

6. Fluid and ion secretion

Due to their diluting and flushing effects, fluid and ion secretion of epithelial cells is another protective mechanism contributing to the barrier function [20]. In humans, the jejunal net water and sodium chloride absorption decreased during both psychological (induced by dichotomous listening) [78] and physical stress (induced by cold pain) [79], and also ion absorption is changed toward secretion in psychological form [78]. Both acute and chronic stress inductions increased short circuit current (an in vitro technique used for measuring the secretory response of intestines) in several parts of rodent intestines [36,41,59,80]. In addition, the peripheral non-selective CRF antagonists astressin or α-helical CRF9-41 abolished stress- induced alterations [41,80,81]. Intraperitoneal injection of a newly developed selective CRF(1) peptide agonist cortagine also induced an increase in defecation and watery diarrhea in mice and rats [82]. The effects of mineralocorticoids and glucocorticoids on intestinal water and ion movements are well known. Methylprednisolone for 3 days increased Na+K+adenosinetriphosphatase activity and Na+ absorption [83]; it also increased guanylate cyclase activity and Cl- secretion in the jejunum and ileum 6 h after administration [84,85]. However, there is no direct information about the increase of water and ion secretion related with increased glucocorticoid secretions in different stressful conditions.

Advertisement

7. Intestinal permeability and mast cell functions

An increase in intestinal permeability was reported in animals [61,86] and humans [87] submitted to acute or chronic stress, and in IBD and IBS patients [88,89]. Increase of intestinal permeability or macromolecular permeability also involves mucosal inflammation, mucosal damage, and mast cell hyperactivity. The barrier properties of the intestinal epithelium are usually studied by assessing the permeability to various probe/marker molecules (such as horseradishperoxidase, 51Cr-EDTA, mannitol) in vivo or in vitro with intestinal segments mounted in Ussing-type chambers [20]. Bagchi et al [90] investigated the effects of acute (90 minute by water immersion) and chronic (15 min/day for 15 consecutive days by water immersion) stress on the production of reactive oxygen species (ROS) and oxidative tissue damage in gastric and intestinal mucosa. Both acute and chronic stress increased ROS production, lipid peroxidation and DNA fragmentation in both gastric and intestinal mucosa, but acute stress produced greater injury when compared to chronic stress. Colonic myeloperoxidase, mucosal mast cell activity and colonic permeability increased (as assessed with macroscopic damage and bacterial translocation to mesenteric lymph nodes, liver and spleen) at 12 weeks-period in maternally deprived rat pups induced by separation from their mothers on 2-14 days period for 3 hours a day [91]. Besides, the responses of maternally deprived rats to the TNBS-induced colitis were more prominent compared with the control rats. Both acute and chronic cold stress could cause oxidative stress of duodenum and a change in iNOS, which was related to the intestinal damage process in broiler chicks [92]. There are also evidences about the bacterial production of iNOS [93]. Boudry et al [58] suggested that both apoptosis in the crypts and an immature epithelium covering the villus surface can be responsible for a barrier defect in rats submitted to WAS (1 h/day) for 5 or 10 days. Morphologic [39,94] and enzymatic alterations [40] in mitochondria of the intestinal epithelium can also participate to promotion of intestinal dysfunctions in stress-induced animals.

Mast cell activity of gastrointestinal mucosa has altered in both acute and chronic stress models. They contain inflammatory and immunmodulating mediators such as prostaglandins, histamine and serotonin that directly alter epithelial transport properties [80] and nerve and muscle functions [95]. So it has a pivotal role in visceral hypersensivity [34], intestinal inflammation, intestinal mucin secretion [96] and epithelial barrier disruption [20,61]. Castagliuolo et al. [96] found that mast cells are essential for the colonic mucin and prostaglandin secretions in immobilization stressed mice, because of these secretions were absent in mast-cell deficient animals. Even, reconstitution of bone marrow with mast cells reversed that response to normal stress values [96]. Mast cell-deficient rats (Ws/Ws) and their normal mast cell-containing littermates (1/1) were submitted to water avoidance (1 h/day) or sham stress for 5 consecutive days. Stress increased baseline jejunal epithelial ion secretion, ionic permeability, macromolecular permeability [61,94], and the number and proportion of mucosal mast cells [94] in 1/1 rats but not in Ws/Ws rats, compared with non-stressed controls. Morphological, inflammatory and permeability changes were not seen in ileum and colon of mast cell-deficient rats in a chronic stress model [42]. Kim et al [97] reported that acute stress increased mast cell number and mucosal proteinase-activated receptor-2 (PAR2) expression (G-protein coupled receptor which can be activated by mast cell tryptase and modulate gastrointestinal functions) in the rat colon. Because of CRF-antagonist astressin inhibited these alterations, they suggested that CRF can be mediator of these events. Dexamethasone treatment improves PAR-2 agonist-induced visceral hypersensitivity, but does not prevent PAR-2 agonist-induced increase in colonic permeability in rats. This effect is coupled with a reduction of colonic mast cell numbers and RMCP-II contents [98]. Chronic stressful stimulus also caused greatest numbers of degranulated mast cells [99] and mast cell hyperplasia in the intestine of rats [100]. In our unpublished study, we found that while acute cold swimming stress (swimming in 18 0C water, for 15 min) increased the mucosal mast cell numbers in ileum, dexamethasone decreased their numbers significantly. It has been reported that mast cell numbers and their protease II activity were decreased in different dexamethasone treatments [98,101,102]. Wrap restraining stress in rats for 2 hours increased histamine content in colonic mast cells without degranulation, and this was found to be mediated by interleukin I and CRF [103]. Santos et al [80] reported that CRH, when injected intraperitoneally, mimicked the effects of acute restraint stress on colon epithelium such as increased colonic ion secretion, macromolecular permeability via cholinergic and adrenergic nerves and mast cells. Because CRH-induced alterations in colon epithelium inhibited by CRH-antagonist, adrenergic, nicotinic and muscarinic receptor antagonists and mast cell stabilizing agent doxantrazole, but not by aminoglutethimide (mineralcorticoid and glucocorticoid synthesis blocker, they suggested that steroids have no role in CRH-induced colonic pathology. They also denoted that stimulatory effects of CRH on mast cells can be mediated by direct or indirect neural pathways. Also in humans, CRH mediates transcellular uptake of HRP in colonic mucosal biopsy samples via CRH receptor subtypes R1 and R2 on subepithelial mast cells [104].

Advertisement

8. Mucin – Physicochemical barrier

Mucin secretion is also a major component of intestinal barrier which protects the mucosa by forming a coating layer over the epithelium against bacterial penetration. In addition to providing a biophysical barrier, mucus forms a matrix that allows the retention of high concentrations of specific and nonspecific antimicrobial molecules, such as secretory IgA and defensins in close proximity to the epithelial surface [23,105,106]. The secreted mucus forms two layers, a thinner inner layer that is accepted to be sterile and difficult to dislodge and an outer layer that is not sterile and is more easily removed. Mainly MUC2 type mucin is synthesized from goblet cells in small and large intestines. Epithelial cells and Paneth cells secrete antimicrobial peptides that help preventing of bacteria to penetrate the inner mucus layer. Both the physicochemical structure and thickness of mucin coating show differences through the gastrointestinal canal. It was suggested that mucus thickness is increased and the increase was correlated with luminal bacterial concentrations of related parts of gastrointestinal canal. The inner layer is ~15–30 μm and the outer layer is 100–400 μm in small intestine and it is thickest in ileum because there are approximately 105–107 bacteria per gram of faeces in the lumen. Otherwise, the inner layer of ~100 μm and a thick outer layer of ~700 μm in large intestine where 1010–1012 bacteria per gram of luminal content resides [106]. Mucin is not only a barrier against the bacteria but also nutritional source for bacteria. In addition, bacteria capable of colonizing mucus can avoid rapid expulsion via peristalsis of the intestine and take an advantage for transmitting their signaling pathway to the host [107]. Microbiota can stimulate mucin secretion via bacterial products and increase MUC2 expression via activation of TLRs and NOD-like receptors or other signaling pathways at transcriptional level. Mucin secretion is also influenced by hormones, inflammatory mediators, several signaling peptides, growth factors and infectious bacteria [106,107]. Castagliuolo et al. [108] reported that acute stress caused a depletion of goblet cells and an increase of mucin secretion related with decrease of mucin containing goblet cell numbers via an increase of CRF secretion [20]. They proposed that although rapid mucin release during acute stress would increase the barrier properties and provide a degree of protection against invasion of a leaky epithelium, goblet cell depletion would be deleterious in a longer time period because of the reduced capacity to respond to ongoing or new threats. A 10-day chronic stress model was resulted in barrier dysfunction in the ileum and colon (increased macromolecular permeability and depletion of mucus) and ultrastructural changes in epithelial cells (enlarged mitochondria and presence of autophagosomes) associated with bacterial adhesion and their penetration into enterocytes [42]. Studies revealed that CRF signaling can activate mucin secretion because goblet cells have CRF1 receptors and stress and peripheral injection of CRF induces mucus depletion in rat distal colon [18]. On the other hand, maternal separation stress increased mucus secretion and thus caused an elevation in the number of mucosal goblet cells in rats [109]. In our unpublished study, both acute cold swimming stress and dexamethasone injection increased the goblet cell counts in the ileum within six hours, but the effects of dexamethasone were more prominent than the swimming stress. Further, while the effect of the dexamethasone on goblet cells maintained in 24 hours period that of swimming stress was disappeared [Ünsal et al., unpublished data]. Finnie et al. [110] reported that exogenous prednisolone and hydrocortisone also increased the mucin secretion significantly in left slightly and in right uninvolved colonic biopsies of patients with ulcerative colitis. They suggested that therapeutic effects of corticosteroids in ulcerative colitis may be related partly with their stimulatory effects on mucin synthesis.

As partly mentioned above, most stress-induced gastrointestinal (GI) dysfunctions can be induced by peripheral CRF agonists and prevented by CRF receptor antagonists [18]. In a review article Larauche et al [18] reported about “CRF signaling” and emphasized that peripheral injection of CRF or urocortin stimulates colonic transit, motility, Fos expression in myenteric neurons, and defecation through activation of CRF1 receptors, whereas it decreases ileal contractility via CRF2 receptors. Additionally, intraperitoneal administration of CRF induces colonic mast cells degranulation via both CRF1 and CRF2 receptors and increases ion secretion and mucosal permeability to macromolecules, which can in turn promote intestinal inflammation and alters visceral sensitivity. Furthermore, CRF peptides can reproduce secretomotor and mucosal alterations in vitro. Although there are a lot of events that CRF is primary mediator in stress-induced alterations of GIS in animals and humans [17,18,32,80-82,111], similar reports for the glucocorticoids are limited [28-30]. Meddings and Swain [28] reported that stress-induced increases in gastrointestinal epithelial permeability seemed to be mediated by adrenal corticosteroids and disappeared after adrenalectomy or pharmacologic blockade of glucocorticoid receptors. Besides, dexamethasone treatment of control animals increased gastrointestinal permeability and mimicked the effects of stress. Spitz et al. [29,30] evaluated the effects of dexamethasone on intestinal barrier functions in various conditions such as starvation and after bacterial contamination. They found that starvation significantly impairs secretory IgA, promotes bacterial adherence to the mucosa and increases intestinal permeability to f-MLP in rats given 0.8 mg/kg dexamethasone intraperitoneally. These effects are significantly attenuated by the feeding of rat chow [30]. In other study, they also found out that dexamethasone administration increased intestinal permeability and bacterial adherence to the mucosa [29]. However, antibiotic decontamination of the intestine completely abrogated the intestinal permeability defects observed in this model. Basing these findings they concluded that bacterial-mucosal cell interactions may be responsible for alterations in intestinal permeability after dexamethasone administration.

Advertisement

9. Intestinal immune system

Intestinal homeokinesis depends on complex interactions between the microbiota, the intestinal epithelium and the host immune system. Innate and acquired immune cells of the intestine have critical roles in barrier function because of they should tolerate the antigens belonging to the food and commensal microbiota as well as they should protect the body against pathogen microorganisms [20,57]. Systemic nonresponsiveness to antigens that are introduced orally is a phenomenon known as “oral tolerance”. Oral tolerance is typically characterized by the suppression of the systemic T helper 1 (Thl) response to antigens and elevated levels of IL-I0, TGF-β and antigen-specific sIgA at the mucosal surface. The T helper 2 (Th2) response also promotes the induction of tolerance in the gut. Production of IL-4 and IL- 5 during Th2 response acts synergistically to enhance IgA production. These cytokines also act further to inhibit the Thl response [57]. The cells of the innate immunity discriminate potentially pathogenic microbes from harmless antigens through pattern recognition receptors (PRR). Toll-like receptors (TLRs) are a family of pathogen-recognition receptors of the innate immune system. TLRs are present on a variety of cell types such as intestinal epithelium, monocytes, and dendritic cells. They recognize conserved molecular motifs on microorganisms called pathogen associated molecular patterns (PAMPS). TLRs are activated by various components of microorganisms, e.g. TLR4 binds lipopolysaccharide (LPS) in gram-negative bacteria. Besides, TLR5 binds to flagellin, TLR2/6 binds to fungal zymosan and TLR7 binds single stranded RNA (ssRNA) from viruses. Activation of the TLRs by either pathogenic ligands or host factors results in downstream activation or inhibition of pathways involved in inflammation. Toll-like receptor activation by commensal bacteria plays an essential role in maintaining colonic homeostasis and controlling tolerance in the gut [57,112,113,114]. However, inappropriate activation of their signaling pathways may lead to deleterious inflammation and tissue injury. TLRs have been implicated in the pathogenesis of many GI disorders [57,114]. Although intestinal immune system is thought to have a critical role in stress-induced alterations of GIS functions, the studies about this situation are limited as this also the case for other functions of GIS such as motility, secretion and permeability [18,115,116]. McKernan et al. [116] investigated for the first time the regulation of TLR expression in the colonic mucosa in two distinct chronic stress models; Wistar-Kyoto (WKY) rats and maternally separated rats where Sprague Dawley rats were used as controls. Significant increases are seen in the mRNA levels of TLR3, 4 and 5 in both the distal and proximal colonic mucosa of MS rats compared with controls. No significant differences were noted for TLR 2, 7, 9 and 10 while TLR 6 could not be detected in any samples in both rat strains. The WKY strain showed increased levels of mRNA expression of TLR3, 4, 5, 7, 8, 9 and 10 both in the distal and proximal colonic mucosa compared to the control animals of Sprague-Dawley strain. No significant differences in expression were found for TLR2 in all samples of both strains. These authors suggested that the up-regulation of TLR 4 and 5 may indicate increases in cytokine production in response to the increases in sensitivity to gut bacteria. In addition, they suggested that the observed differences in TLR expression activity between MS and WKY rats might be related with their different neuroendocrine responses or microbiota. In spleens isolated from mice subjected to chronic 12-hour daily physical restraint for two days, TLR-4 expression significantly increased, T helper 1 (Th1) cytokine IFN-γ and IL-2 levels were found to be decreased, but Th2 cytokine and IL-4 increased. They suggested that stress modulates the immune system through a TLR4-dependent mechanism, because TLR4-deficient mice are resistant to stress-induced lymphocyte reduction and the restraint stress significantly inhibits changes of Th1 and Th2 cytokines in TLR4-deficient mice compared with the wild type mice [117]. Repeated social defeat stress (SDR) has been shown to increase the expression of TLR2 and TLR4 [118] and can activate dendritic cells for enhanced cytokine secretion in response to TLR specific stimuli. Besides, glucocorticoid resistance was determined in CD11+ dendritic cells isolated from spleens of SDR mice, whereas under baseline conditions DCs are highly sensitive to glucocorticoids [119]. Glucocorticoids and catecholamines appear to be able to regulate the expression of certain TLRs [116]. Toll-like receptor agonist-induced cytokine (IL1b, IL6, IL8 and TNF-α) release was markedly enhanced in stimulated whole blood samples from IBS patients compared with healthy controls. Plasma levels of cortisol, IL-6 and IL-8 were also significantly increased in IBS patients [120].

Chronic stress (induced by water avoidance stress for 1 hour/day for 10 consecutive days) induced the infiltration of neutrophils and mononuclear cells, and increased myeloperoxidase (MPO) activity in ileum and colon mucosa, but these changes were not shown in mast cell deficient rats [42]. Similary, jejunal inflammatory cells such as neutrophils, eosinophils and mononuclear cells and expression of IL-4 (TH2 type cytokine) increased, while interferon–γ (IFN- γ) (TH1 type cytokine) decreased in same chronic stress model of rats. Treatment of stressed rats with an antagonist to CRH eliminated the manifestations of intestinal hypersensitivity [121].

Velin et al [122] evaluated the M cell-containing follicle associated epithelium (specialized in antigen uptake) in acute and chronic stress conditions. Acute stress increased horseradish peroxidase (HRP) flux in villus as well as in follicle-associated epithelium (FAE), and chronic stress increased E. coli passage in follicle-associated epithelium whereas there was no significant increase in villus epithelium. In patients with Crohn’s disease (CD), transmucosal uptake of non-pathogenic E. coli across the FAE increased in ileum, despite unchanged macromolecular permeability, but these changes were not observed in patients with ulcerative colitis [123]. Recently, Keita [124] showed that application of CRF agonist increased HRP and E. coli passage, stress-induced increases in uptake across FAE of HRP, and E. coli were reduced by CRF antagonist, mast cell stabilizer and atropine. Chronic restraint stress increases eosinophils expressing CRF in the jejunum, which participate to the recruitment of mast cells and epithelial barrier dysfunction [125]. The influence of CRF signaling on intestinal mast cell activity is detailed above. However, the information about their effects on other immune components of intestine (such as intestinal epithelial cells, TLR expression or intraepithelial lymphocytes) is limited. Larauche et al [82] reported that treatment of mice with CRF-1 agonist cortagine exhibits a dose-related interferon-γ (IFNγ) response indicating T cell and/or natural killer (NK) cell activation, which is followed by tight junction deregulation and dose dependent apoptotic loss of different cell populations in ileum.

IECs participate in initiating adaptive immune responses in the gut by transporting luminal antigens to underlying immune cells for presentation by professional antigen presenting cells or can present antigen themselves [57]. intraepithelial lymphocyte (IEL) and paneth cells (specialized IEC located at the base of intestinal crypts in small intestine) do also synthesis the antimicrobial peptides such as lysozymes, alpha defensins, cathelicidins, lipocalins, and C-type lectins such as RegIIIγ [126]. Production of RegIIIg [127] and alpha-defensins [128] as well as that of secretory IgA [129] are induced by commensal bacteria. Nutritional and infection stress affected the secretory activity of Paneth cells in human [130]. In women, acute cold stress induced the release of α-defensin in the jejunum [131]. Evidences suggested that dysfunction of Paneth cells and impaired defensin secretion may contribute to IBD susceptibility [57,105].

Corticosteroids and catecholamines are well recognized and accepted powerful regulators and players of the body in its response to environmental challenges including biological factors of stress. Stress or corticosteroid applications are known to have also profound effects on intestinal wall structure and functioning. Intestinal submucosa is the place where lymphocytes, eosinophils and mast cells reside in men and animals under normal conditions. Jarillo-Luna et al. [115] investigated the effects of chronic restraint stress in mice submitted to different procedures (adrenalectomy, chemical sympathectomy, and treatment with a glucocorticoid antagonist (RU486), dexamethasone, and epinephrine) on intraepithelial lymphocyte (IEL) numbers. They found that chronic restraint-stress reduced the IEL population in the small intestine and adrenal catecholamines and glucocorticoids are essential in preserving IEL population because adrenalectomy, treatment with RU-486 and chemical sympathectomy decreased the number of γδ, CD4+ and CD8+ T cells in non-stressed groups. They also found that adrenalectomy did not buffered the stress-induced reduction in CD8 lympocytes, but glucocorticoid receptor antagonist RU-486 buffered stress-induced decrease in γδ and CD8+, but not in CD4+ T cells. Besides, low and high doses of dexamethasone (5 and 50 mg/kg BW) significantly reduced the number of γδ and CD8+ T cells, and epinefrin (0.1-0.5 mg/kg) reduced the number of γδ, CD4+ and CD8+ T cells in intact mice. Also many other studies reported that both stress-related endogen rises of glucocorticoids [115,132-134] and exogenous glucocorticoid administrations [135,136] induce decreases in intraepithelial lymphocytes and/or those in ileal Peyer patches. Pretreatment with glucocorticoid receptor antagonist mifepristone significantly reduced apoptosis in both T- and B-cell populations in intraepithelial lymphocytes after the burn injury [133]. Experimental studies also suggested that single or repeated parenteral applications of cortisone cause a decrease in eosinophil concentration in all parts of examined gastrointestinal wall from stomach up to colon [137,138]. Immunosuppressive effects of glucocorticoids are explained mainly by an increase of apoptosis and a decrease of cytokine production. Corticosterone impaired the maturation of DCs and cytokine production and reduced the ability of DCs to prime naive CD8+ T cells in vivo; there was no reduction in surface TLR4 expression in CORT-treated DCs [139]. However, McEwen et al. [140] proposed that although glucocorticoids are mainly known with and used widely for their immunsuppressive aspects, adrenal steroids play also different roles as important modulators of the immune system. Pharmacological as well as physiological changes in glucocorticoids result in a decrease in lymphocyte, monocyte and eosinophil numbers and an increase in neutrophil numbers in the blood of men and rodents. These changes are not related with the glucocorticoid-induced leukocyte deaths. The nonspecific stress and glucocorticoid administration cause redistribution of leukocytes from peripheral blood into various tissues and organs, such as bone marrow, spleen and lymph nodes [140-142].

Advertisement

10. Intestinal microbiota

One of the important groups of the environmental biological stressors belongs to the microbiota, and it is associated with every multicellular organism on earth. It is estimated that in humans and many animals reside at least 1014 microorganisms, making approximately 1.5 kg biomass, in various parts of the body, most abundant of them residing the distal part of the gut in humans and animals with exception of ruminants [143-146]. These parts of the body lined by the skin or mucous membranes and all are in direct contact with the environment. Although the microorganisms are preferable grouped as pathogenic, while they harm the multicellular organisms, and saprophytic, while they seemingly do not harm their hosts instead they build symbiotic relationships, but possibly this depends only on the balance or imbalance among numerous groups of microorganisms constituting the microbiota in a definite part of the body and between the microbiota and its host, in general.

The first days, weeks, months or years, the time spent in mothers’ womb or in egg-shall of the metazoan life is the only time which they are free of microbes. The delivery into the outside exposes them to an enormous range of microbes from diverse environments. This is the first encounters with life forms with different morphology and functions. The studies have shown that within a short time following delivery, the microrganisms are present on the skin and mucosal surfaces of the body. With time, a dense, complex gastrointestinal microbiota develops [147,148]. Due to the unique properties of microorganisms including their small size, metabolic versatility and genetic plasticity, microorganisms can tolerate and easily adapt to unfavorable and immense variety of continuously changing environmental conditions [149]. However, although a wide variety of microorganisms were exposed to representative individuals from start up throughout the life periods, only a limited numbers of species are able to colonize permanently in available body surfaces of man and animals. The microorganisms display a tissue tropism; e.g., they colonize predominantly only certain body sites. Consequently, each side is inhabited by only certain species of microorganisms. The microorganisms found at a particular body site constitute what is known as the indigenous or normal microbiota of this site, wherein the term ‘indigenous’ include all of viruses, protozoa, archae, and fungi [150,151]. The GIT is inhabited with 1013–1014 microorganisms, approximately 500 to over 1000 different species and more than 7000 strains. Their counts exceed ten times the numbers of somatic cells of their hosts [144,151-154].

The very complex and diverse gastro-intestinal microbiota differs from species to species, in dependence of nutritional habits with some geographic motives. Besides, it varies from one segment to another and varies over time in the same individual, because the environment of the GIT varies considerable along its length and with the lifetime of an individual [144]. Thus, the composition and intensity of the microbiota of a newborn is quite different from those of an adult which are in turn quite different from that of an elderly individual [155,156]. Colonization begins at birth with facultative bacteria and the colonization of anaerobic bacteria which are composed of more than % 90 of GIS microbiota develops later. In humans, the microbiota has a stable adult-like signature by 1 year of age [113,154]. Similarly, the composition and intensity of the microbiota varies along the gastrointestinal tract where they are attached to the mucosa or are present in the contents. In stomach and duodenum of humans, microorganism numbers is 103CFU/ml and include more lactobacilli, streptococci and yeasts species. In jejunum (104CFU/ml) and ileum (107-8 CFU/ml) Lactobacillus, Bacteroides, Enterobacteriaceae, Streptococci, Bifidobacterium and Fusobacteria species are more existent. The highest numbers of bacteria (1011-12 CFU/ml) displaying enormous diversity are found in colon, predominant species being Bacteroides, Fusobacterium, Eubacterium,Peptococcus, Peptostreptococcus, Veillonella, Bifidobacterium, Escherichia, Clostridium, Lactobacillus and others [147,154]. Although only 40-45% part of GI microbiota could be growth with classical microbial culture tecniques, recently developed moleculer techniques allow the definition of non-culturable members of microbiota. The vast majority of microorganisms belong to the phyla of Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria while Fusobacteria, Verrucomicrobia and TM7 are represent in relatively lower numbers [144,145,157,158]. There are also some fungi and Archaea present in the gastrointestinal tract [144,159,160], but they comprise less than 1% of total inhabitants. The majority of the intestinal bacteria are composed of gram-negative anaerobes [143].

With the acquisition and establishment in the intestinal lumina with various microbial populations bidirectional interactions between microbiota and host organism starts [161,162]. There are symbiotic relationships between the host and its GI microbiota in steady-state conditions. The gastrointestinal tract serves a natural habitat for a dynamic microbial community of different origins while the microbiota is very essential for both gastrointestinal integrity [163] and general health of host organisms [113]. Comparison between the conventional and germ-free animals has allowed obtaining information about the effects of microbiota on morphologic, functional and metabolic characteristics of host organisms [23,113,154,160]. Germ free animals have enlarged cecum [164], they have thinner gut wall, smaller total surface area and more cubic epithelium than the columnar, increased enterochromaffin cell area and smaller Peyer’s patches, and intestinal epithelium has slower turnover rate compared with the conventional animals [160,165]. The materiality of the gut microbiota also for many gut functions such as motility, immune and barrier functions are reviewed by several authors [23,113].

Complex association between the host and its microbiota collectively extends the processing indigestible parts of food to the benefits of the host organisms via metabolic capacities which are not coded in host genomes like mammalian and avian species [166-168]. They digest the unusable parts of the diets; metabolites reach to the intestinal lumina within various secretions and desquamated cells of their hosts for growth and proliferation. At the same time, they also supply the host organisms with a considerable amount of nutrients, which makes this ecosystem an invaluable, essential metabolic organ, which contributes significantly to the homeokinesis of the host organisms [23,113,152,169]. They produce substances including vitamins, volatile or short-chain fatty acids (SCFAs) and polyamines which are absorbed throughout the intestinal wall and used directly by intestinal epithelial cells or other cells of the body [170,171]. SCFA profoundly influence gut barrier functions, host immunity, epithelial proliferation and bacterial pathogenesis [172]. Zheng et al. [171] showed extensive gut microbiota modulation of host systemic metabolism involving short-chain fatty acids, tryptophan and tyrosine metabolism, and possibly a compensatory mechanism of indole-melatonin production. All these metabolites have also many regulatory functions in host organisms. Tryptophan and tyrosine are precursors of neurotransmitters acting directly at the central nervous system level [113]. Thus, the gut microbiota enhances the host’s metabolic capacity for processing nutrients and modulates the activities of multiple pathways in a variety of organ systems, including the brain.

Microbiota acts as a luminal barrier against incoming pathogens; this phenomenon has been described as colonization resistance [23,154]. Beneficial and pathogen microorganism compete with each other for the attachment sites and for nutrients, so microbiota and their products can prevent pathogen colonization directly. On the other hand, microbiota acts on barrier functions also indirectly by stimulating mucosal immune system [172]. The abundant antigenic stimulus supplied by microbiota and their products are essential for the stimulation of immune system cells locally and systemically [173-175]. In germ-free animals, besides the poor developed Peyer’s patches, altered compositions of CD4+ T cells and IgA-producing B cells in the lamina propria [165], TH 17 cells, which is a subset of the T cells and contribute to resistance against colonization by pathogens were virtually absent in germ free animals [176,177]. Chow and Mazmanian [177] denoted that although Th17 cells are essential for immunity, they have also been implicated in the pathogenesis of many autoimmune diseases, including IBD, arthritis, psoriasis, and experimental autoimmune encephalomyelitis (EAE).

The gut microbiota has recently been identified as the main source of highest biological variability confined in an individual [178]. Because the metazoan life-forms and the inhabitation of their gastrointestinal tract with microorganisms evolved together, there are close links between any host or its epi-genome and its very complex diverse gastrointestinal microbiota with their multitude genomes. It is estimated that this microbial community has 70–140 times more total genes than the human host. These functional inter-relationships between host and microbiota or two different genomes determine the health or disease state of the metazoan hosts and the balance among different microorganism populations [113,172,179]. It is well accepted that the intestinal microbiota involves in metabolome of the host, thus promotes actively fat accumulation and weight gain and sustains indirectly a low-grade systemic inflammation especially when imbalanced, and consequently, enhances the risk for complex, multifactorial diseases such as insulin resistance, diabetes, obesity and cardiovascular diseases. The search of global obesity epidemic led to the growing evidences about the possible roles of intestinal microbiota in these respects [180,181,182]. Claus et al. [170] has noted that the colonization of the gut microbiota was associated with a rapid increase in body weights of animals up to 4% within 5 days of colonization. Findings of various studies also revealed that gut microbiota profile of obese and diabetic individuals differ by phylum level both in its quantity and quality from that of lean and nonobese individuals [182,183]. Recent evidences exhibit that the composition of the gut microbiome may influence body weight of the host by various mechanisms including enhancing the ability of intestines to extract energy from food [184], regulating fat storage in tissues [185] and affecting satiety by modulating the levels of local hormones that regulate satiety and by direct effects in central nervous system [186].

There is a growing appreciation of the critical roles played by the commensal microbiota, both in general well-being of the hosts and in the specific functioning of the brain–gut axis [113]. Bidirectional communications of brain–gut–enteric microbiota axis simply actualized by through signals from the brain can influence the motor, sensory, and secretory modalities of the gastrointestinal tract and conversely, visceral messages from the gastrointestinal tract can influence brain functions [187]. Sudo and colleagues [25] showed that gut microbiota effect the stress responses of host organisms. They compared the response of the HPA axis to stress in GF, specific pathogen free (SPF) and gnotobiotic mice that were mono-associated with a single bacterium. Restraint stress caused an exaggerated ACTH and corticosterone elevation in GF rather than SPF mice. This hyper-response of the HPA axis was reversed by mono-association with Bifidobacterium infantis. They also showed in following experiments that the levels of brain-derived nerve factor (BDNF), norepinephrine and 5- 5-hydroxytryptamine (5-HT) in the cortex and hippocampus were significantly lower in GF mice than in SPF mice [188]. Improvements of stress-related symptoms by probiotic administration also support the possible regulatory effects of microbiota on HPA axis and brain functions [44,189]. Gareau et al [44] reported that probiotic treatments improved colonic dysfunction and corrected the higher corticosterone levels in stressed rats induced by maternal seperation. L. rhamnosus (JB-1) reduced stress-induced corticosterone and anxiety- and depression-related behavior in mice. In different region of brain, this therapy also altered GABA receptor expression implicated in the pathogenesis of anxiety and depression, which are highly comorbid with functional bowel disorders. Due to the neurochemical and behavioral effects were not found in vagotomized mice, they suggested that vagus could be a major modulatory constitutive communication pathway between the bacteria exposed to the gut and the brain [189]. The roles of probiotics and gut microbiota in modulation of visceral and even somatic pain perception and their possible roles in alterations of mood and behavior were reviewed by Forsythe et al. [190] and Grenham et al. [113].

As mentioned above, intestinal dysbiosis can adversely influence gut physiology both by direct effects to the surrounding gut wall and by leading to inappropriate brain–gut axis signaling and associated consequences for CNS functions and disease states. Stress at the level of the CNS can also impact on gut functions and lead to perturbations of the microbiota [113].

11. Stress and intestinal microbiota

Intestinal microbiota have once been seen as potential treat for the host organisms, but recently accepted as an integral part of metazoan life and even as an organ with a huge variety of building blocks which mainly cooperate with each other and with the host for maintaining the health and survival [113,144]. However, various stress factors such as heat, cold, nutritional alterations, overcrowding, physical restraints and transporting or fouled or contaminated foods can destroy the microbial balance in the gastrointestinal system [66,67,191-195] and alter their relationships with each other and with their hosts. Stressful stimuli can affect gastrointestinal microbiota directly, for example via limited availability of food ingredients or direct actions of stress mediators such as adrenaline or noradrenaline on microbiota [27,196], and indirectly via altering the intestinal environment of bacteria such as intestinal secretion, motility, permeability and immune functions as reviewed above.

The effects of several stressors and stress mediators on intestinal microbiota were given in Table 1. Bailey et al [197] induced social disruption stress (SDR) in mice to determine whether the microbiome contributes to stressor-induced immune enhancements. They analyzed bacterial populations in the cecum with using bacterial tag-encoded FLX amplicon pyrosequencing (bTEFAP) and found that microbiota significantly changed immediately after stressor exposure as summarized in Table 1, and stress also increased circulating levels of IL-6 and MCP-1, which were significantly correlated with stressor-induced changes to three bacterial genera (i.e., Coprococcus, Pseudobutyrivibrio, and Dorea spp). However in antibiotic treated mice, exposure to SDR failed to increase IL-6 and MCP-1. Bailey et al [191] reported that restraint stress also significantly change the composition of the intestinal microbiota in mice and disruption of the microbiota increased susceptibility to murine enteric pathogen Citrobacter rodentium which can be associated with reduced competitive exclusion of commensal bacteria and increased tumor necrosis factor alpha (TNF-α) gene expression in colonic tissue. Reduction in bifidobacteria and lactobacilli numbers in fecal samples of infant monkeys whose mother had been exposed to stress in either early or late pregnancy period showed that maternal pregnancy conditions affect infant health and can enhance susceptibility to infection [68]. Similarly, early life stress induced by maternal separation altered fecal microbiota with concomitant increases in corticosterone and also visceral hypersensitivity and systemic immune response to in vitro lipopolysaccharide challenges in rats [198]. Knowles et al [199] reported that non-extreme ‘every day’ stress events such as exam stress can affect the integrity of the indigenous gastrointestinal microflora of humans but these changes are not supported by cortisol responses. Maternal separation of rhesus macaques also caused to decrease of lactobacilli at 3th days post-separation but significant differences in the cortisol responses did not predict the magnitude of the reduction of lactobacilli numbers. These authors suggested that more than one neuro-hormone can modulate microbiota changes.

Because of their immune-suppressive or stress-mediating effects, some studies have been focused on the effects of glucocorticoids on intestinal microbiota. These studies showed that exogenous glucocorticoid applications to the host organisms are also able to cause changes in gastrointestinal microbiota. We [200,Ünsal et al., unpublished study] and others [29,30,201] demonstrated that exogenous glucocorticoid administrations can also affect gut microbiota by enhancing total aerobe and gram negative enteric bacteria and their translocation to extraintestinal tissues [201]. We compared the effects of different doses of dexamethasone on ileal microbiota and found, in contrast to well-known stress effects, that 5mg/kg dexamethasone injection also increased the numbers of total anaerobe and lactobacilli in ileal content of rats [200]. However, their number did not change in lower doses of dexamethasone. Also in our unpublished study acute cold swimming stress decreased the numbers of lactobacilli, while dexamethasone in dose of 5 mg/kg increased total aerobe, gram-negative enteric bacteria and lactobacilli. Thus, the evidences available suggest that several stressors reduce the number of lactobacilli, while on the contrary, they increases growth, epithelial adherence and mucosal uptake of Gram-negative pathogens. Lactobacilli may possible be defined as stress indicator bacteria of the gut microbiota which is sensible to the effects of various stressors, in general. Although it is known that exogenous glucocorticoids increase the counts of gram negative enteric bacteria [29,30,201], no other information about their effects on the lactobacilli in the gut could be found.

The roles of stress and stress-related hormones in the pathogenesis of infectious diseases are beyond any argument. Microbial endocrinology is a new research area which appeared from the demand how stress influence the bacterial infections, how neuro-endocrine-immune secretions of host organisms influence their harboring microbiota and how infectious microbes can actively use the neurohormonal products of the stress to their own advantages [202,203]. Recently, several in vitro studies have focused on direct effects of stress hormones on bacterial growth and their virulence in an effort to explore and understand the interactions of so-called stress hormones and infections. These studies gathered the evidence that catecholamines stimulate the growth of a wide variety of gram-negative bacterial species, including those of medical importance [27,196,204-209]. Furthermore, catecholamines were also found to be able to induce E.coli to produce a heat-stable autoinducer of growth [19,27,204,210] as well as for adhesion required K99 pilus and shigella-like toxins I and II, which may have important roles in its pathogenic activity [210].

The effects of norepinephrine and its receptor antagonists on mucosal bacterial adherence were also determined in sheets obtained from different parts of the gut, mounted in Ussing chamber. Norepinephrine increased the adherence of enterohemorrhagic Escherichia coli O157:H7 (EHEC) to colonic epithelium through interactions with α-2 adrenergic receptors [211-213] and it increased the internalization of enterohemorrhagic E. coli O157:H7 (EHEC) and S. enterica serovar Choleraesuis in jejunal mucosa containing Peyer’s patch follicles [214,215]. Parallel studies related to the direct actions of other stress mediators such as cortisol or CRF on bacterial growth could not be found. However, Kakuno et al. [216] suggested that hydrocortisone enhance intracellular colonizations of E. coli and Schreiber and Brown [217] reported that ACTH increased EHEC adherence to the porcine colonic mucosa.

Many factors participate in the pathology of inflammatory bowel disease (IBD) such as genetic and immune status of host organism, the gut microbiota, and environmental triggers [218]. Some scientific events support that the enteric microbiota is involved in abnormal inflammatory responses observed in diverse animal models for inflammatory bowel disease [219,220]. These events reviewed by the authors [219,220] are arranged as the reduction or absence of intestinal inflammation in animal models of colitis in antibiotic-treated or germ-free animals; colitis formation with some bacterial inoculations to germ free rats or not to formation with other bacterial species; and beneficial effects of probiotics and prebiotics in IBD patients or animal models of IBD. However, although microbiota has been thought to play an active role in etiopathogenesis of inflammatory bowel disease, it is not clear whether they are cause or outcome. In other words, whether alterations in intestinal microenvironment of microbiota such as mucin secretion, immune modifications and epithelial dysfunctions cause to changes in commensal microbiota or microbial alterations cause to inflammatory responses of intestinal mucosa in stressful conditions is not clear yet [221].

Although the reports in microbiome composition of IBD-patients or models show differences, changes in microbiota composition are characterized more likely by decreases in lactobacilli and bifidobacteria, and increases or unchange in aerobe and facultative anaerobic bacteria [222]. Molecular analysis of the microbiota of IBD patients have shown that temporal stability and diversity of the gut microbiota composition in IBD patients decreased compared to non-IBD controls [223], and commensal bacteria, particularly members of the phyla Firmicutes and Bacteroidetes decreased and Proteobacteria and Actinobacteria increased [224]. These changes look alike to those that seen following effects of stress on intestinal microbiota.

Stress and/or Stress MediatorsMicrobiotaMethodReferences
Social disruption stress (SDR) in mice (total of 6, two-hour cycles of SDR)Decrease in Bacteroides spp., tended to decrease in Lactobacillus spp., tended to increase in Clostridium spp. and changes in Coprococcus, Pseudobutyrivibrio, and Dorea spp. in cecal content(bTEFAP) 197
Restraint stress for 7 days in mice
(between 18.00- 08.00 h)
Overgrowth of facultatively anaerobic microbiota, reduction in family Porphyromonadaceae,and genus Tanneralla,reducing microbial richness and diversity in the cecaboth culture technique and bTEFAP 191
Prenatal stress induced by acoustical startle paradigm
for 6 weeks in early or late pregnancy in Rhesus Monkey
prenatal stress reduced the overall numbers of bifidobacteria and lactobacilli in fecal cultures of infants culture technique68
Maternal separation in rhesus macaques (Macaca mulatta)Decrease of lactobacilli in fecal samples at 3th days postseparation, not correlated with cortisol responses culture technique26
Academic stress in undergraduate studentsReduction in fecal lactic acid bacteria, unsignificantly cortisol enhancement culture technique199
Food, water and bedding deprivation in mice Decrease in lactobacilli in stomach, increase of coliforms in jejunum, ileum and cecum, reduction in fusiform-shaped bacteria associated with mucosal epithelium of cecum and colon culture technique192
Dexemethasone (0.8 mg/kg)
Dexamethosone (0.8 mg/kg)+ starvation for 48 h in Fischer rat
impairs secretory IgA, promotes bacterial adherence to the mucosa, increase of intestinal permeability 29,30,225
Dexamethasone 5 mg/kg in ratsIncrease of total aerobe bacteria and lactobacilli in ileum culture technique200, Ünsal et al., unpublished study
Methylprednisolone (3 mg/kg) in rats subjected to temporary liver inflow occlusionIncrease of intestinal Klebsiella spp and Proteus spp and of translocation to multiple organs.culture technique 201
Pregnant rats were treated with either cortisone acetate or normal saline on days 18-21 of gestationTotal bacteria and gram-negatives found in association with the mucosa were significantly lower in pups prenatally treated with steroids. culture technique226
Norepinephrine action on porcine or murine cecum/ colon/jejunum explants Increases cecal-colonic adherence of E. coli O157:H7; changes Salmonella and E. coli uptake into Peyer’s patches211-215
ACTH action on explants of porcine distal colonic mucosa Increases adherence of E. coli O157:H7 to colonic mucosa217

Table 1.

The effects of some different type stressors and/or stress mediators on intestinal microbiota

12. Nutritional stress

Nutrition and nutrients of metazoans play very important multifunctional key roles both for metazoan host and for its gastrointestinal microbiota. They are essential not only in colonization, growth and survival of the microbiota in intestinal system but also in maintaining the balance among different species and their localization within its lumina [67,192,227,228]. Nutrition plays an important role as stressor for host organisms and their gastrointestinal tract via three mechanisms. Firstly, foods supply nutrients for intestinal microbiota and also serve as carriers of various microorganisms into gastrointestinal tract which may under circumstances lead to imbalances among different species; secondly, nutritional deficiencies or imbalances are perceived from the organisms as stressors setting them to the state of well-known alarm reaction of stress; and thirdly, certain food ingredients in their undigested forms as foreign substances accepted as non-self from organisms and stimulate a stress situation. In cases of carrying the microorganisms into gastrointestinal tract or acting as antigens, the foreign treats come into direct contact with the wall of gastrointestinal tract. A great part of microorganisms stay in gastrointestinal canal for a short time period, while others colonize its lumina permanently and their genera can be life-long present there, mostly in a symbiotic relationship with the host organisms. Dietary ingredients, which are not digestible for the host organisms or which are digestible but escape from the intestinal digestion can be utilized as substrate for growth from microbiota colonized in the following sections of the gut [229,230]. All microorganisms residing in gastrointestinal tract needs the nutrients that necessary for their growth and proliferation are continuously supplied via foods of their hosts and from host organisms in secretions of digestive organs including saliva, gastric juice, pancreatic, hepatic (bile) and intestinal wall secretions and desquamated epithelial structures. The composition and amount of the food, even the compositions of these secrets may undergo substantial changes. This is why the hosts’ balanced nutrition and physiological state exerts a strong controlling effect on its microbiota [160,181,192,231-237]. Thus, any nutritional deficiency or imbalance of their hosts serve the most important challenge for the growth, survival and balance of different microbial species within the intestinal lumina.

Malnutritions in different nature or nutritional imbalances had always been and are still very widespread health problems of men and animals worldwide and frequently seen in infants and elderly, and those subjects having malignancies, getting chemotherapy and/or radiotherapy or infected with human immunodeficiency virus. Very common forms of malnutrition are protein, calorie and protein calorie deficiencies which almost always are complicated with deficiencies of other nutrients, especially that of minerals and vitamins [238]. As mentioned above, deficiencies of calories, proteins, minerals or vitamins in hosts’ food can influence the indigenous microbiota both directly via the restricted availability of the metabolites and their indigestible parts for hosts and indirectly by inducing a stress response within the host organism and affecting the compositions of gastrointestinal morphology and secretions as well as by impairing the general and local immune responses and neuro-endocrine-immune network leading to an imbalance between the host organism and its microbiota, in general. These changed milieus offer possibilities to certain new species of the microbiota for adaptation in gastrointestinal tract or cause their dispersion from their localization areas to others. In protein calorie malnutrition, colonic type microbiota known to spread to and proliferate in the upper small intestine which may cause a variety of metabolic disturbances including steatorrhea, vitamin deficiencies, nutrient malabsorbtions, and consequently water leakage into lumina and diarrhea [160,239-241]. Generally, pathogenic microorganisms including Enterobacteriaceae, Pseudomonas, Klebsiella and Candida were increased [240,241]. In such clinical cases of complicated protein and/or calorie malnutrition, pathogenic microorganisms often cause endotoxemia and infection in addition to intestinal disruptions including diarrhea and metabolic diseases [242,243]. Thus, they all affect the composition of the intestinal digesta and its passage time, which in turn may influence the composition of the indigenous intestinal microbiota and their relationships with the intestinal wall [244-246].

Generally, experimental studies use deficiencies or excesses of a definite dietary component or several components, and animals are held under more hygienic, defined conditions throughout the rearing and experimental periods than their counterparts, whereas clinical cases develop spontaneously in man and animals. Thus, they give the possibility to detect the possible effects of a certain dietary component on the behavior of the gastrointestinal microbiota. However, such experimental evidences from studies using deficiencies or imbalances of definite nutrients in this respect are very sparse. So, an almost protein-free diet disrupted the cecal microbiota, and made mice more susceptible to bacterial translocation than those mice nourished adequately [227,236,247]. The counts of cecal total aerobic bacteria and Gram-negative enteric bacilli were found to be increased time-dependently when CD-1 mice were fed an almost N-free diet for 21 days [236]. The results of another study on adult female Crl:CD-1[CR]BR mice also showed that both the feeding an almost protein-free and 20% fat containing diet for 14 days and starvation for 3 days resulted in an increase in counts of Gram-negative enteric bacilli and a decrease in counts of lactobacilli and strict anaerobes [227]. In certain studies the effects of dietary manipulations combined and/or compared with those of endotoxemia were investigated. So, Deitch et al. [227] studied the effects of starvation and malnutrition alone or in combination with endotoxemia and found that the spread of bacteria from the gut could not be controlled nor translocated bacteria be cleared in protein malnourished mice as effectively as in the controls. However, no association between protein malnutrition and bacterial translocation could be found by Katayama et al. [247]. Instead, these authors determined that the total numbers of Gram-negative enteric bacilli adherent to the mucosa of ileum and cecum were less in protein malnourished rats than in their adequately nourished controls. Further, there was also a significant negative correlation between the duration of protein malnutrition and bacterial adherence to the intestinal mucosa. Only, E. coli binding to insoluble ileal mucus was increased in the rats receiving endotoxin. Tannock and Savage [192] reported that the deprivation of food and water and bedding for 48 hours increased the counts of coliform bacteria while they decreased the counts of cecal lactobacilli of CD-1 and C57BL mice strainsIn a preliminary study, we found that feeding an almost protein-free diet to male Wistar rats for 35 days affected especially the total aerobe microorganisms and lactobacilli while total anaerobe and Enterobacteriaceae remained relatively unaffected. Compared to controls with balanced nutrition, both dietary qualitative and quantitative protein malnutrition decreased mean lactobacilli counts. Also, the quality and quantity of the dietary protein made a difference in their effects on intestinal microbiota; compared to gelatin-fed animals, lower aerobe and higher lactobacilli counts could be observed in cecal samples of rats given an almost protein-free diet. Furthermore, it could also be shown that the actual immune status (e.g. suppression of neutrophils) of the host can modify the effects of the qualitative and quantitative protein malnutrition on the intestinal microbiota [193].

Human cultural characteristics may also have implications on the compositions of the gastrointestinal microbiota. Living on a high carbohydrate diet caused also the presence of fewer bacteriodes and more enterococci in feces of the people than those living on a Western diet with more fat and animal proteins [231].

After all, the mechanisms via which different type of diets or dietary manipulations affect the host and its guest organ ‘gut microbiota’ are still not exactly cleared. The effects of dietary qualitative and/or quantitative protein malnutrition on regulatory systems in men and animals are well characterized and are the topic of numerous texts. Earlier studies with definite protein malnutrition were summarized by Aschkenasy [248] and suggest that protein malnutrition of different types or amino acid imbalances generally result in increases of adrenalin and glucocorticoid concentrations in man and animals. Torún and Viteri [249] also noted that in protein and/or protein-calorie malnutrition, the concentrations of adrenalin and glucocorticoids are either increased or showed no important change while many other hormones with exceptions of aldosterone and growth hormone decreased significantly. However, such studies have very important drawbacks as they look only one aspect of the regulators such as their concentrations in blood. Generally, the concomitant expression status of the enzymes which interconvert active and inactive forms of a given hormone and their receptors in target tissues or cells are ignored or not evaluated concurrently. A study conducted by Marroqui et al. [250] on mice demonstrated that during a protein malnutrition plasma glucagon concentration increased, but the ability of exogenous glucagon to raise plasma glucose levels were lower in mice given a low protein diet.

13. Conclusion

Since their introduction in the terminology of scientific medicine, the terms environment, stress and microorganisms were probably never been so important in mind of mankind for the development, health and welfare of men and animals. Although the roles of stress and stress-dependent disruptions of the intestinal microbiota both in developments and in promotion of the symptoms of various diseases and disorders including those of gastrointestinal system in men and animals are well accepted, there is still a lack of information about many aspects including which strains play really a role in the etiopathogenesis of a given condition, and which mechanisms are effective in such cases [24,113]. The stress-dependent dysfunctions of HPA axis can manifest itself in different ways. In many cases it may be related to high or low cortisol concentrations in blood whereas in other situations no detectable change of cortisol occurs. Further, the response given by hypothalamus and pituitary gland to the cortisol can be increased or decreased depending on the receptor numbers [10,11,14,15]. While in classic stress response sympathetic nervous system and glucocorticoids thought to be responsible for stress-dependent processes, studies within last two decades suggest that many disordered situations of the gastrointestinal system mediated by CRF. Both CRF-related peptides and CRF receptors are also expressed within the intestine, where they may activate directly the enteric, endocrine, and immune cells and may be involved in intestinal manifestations such as mucosal permeability, secretion, mast cell function, motility, mucin formation, immune function and many disorders of the gut. In other words, the peripheral changes produced by stress can be mimicked by CRF-injection and prevented by CRF-receptor antagonists [18,20,31,32,82]. Therefore, CRF have been suggested as a new target in treating stress induced functional gastrointestinal disorders.

Recently, intestinal microbiota imbalances gain growing interests both as the subject and cause of stress and stress-related diseases which are connected with not only the gastrointestinal system but also all other systems or organs of the metazoan hosts including the adipose tissue [24,113, 179, 184,185]. Basing on experimental and clinical studies, certain phyla and species are currently related with a given specific condition [180,181]. However, all these studies are looking mainly on one side of the iceberg, like for example changes in a specific member of the microbiota in respect to stress stimuli and a specific neurotransmitter in brain, as it also the case in search of the mediating regulatory pathways. Understanding the roles of stress and stress-related microbiotal changes and their mechanisms in the role of various physiopathological conditions would be helpful in improvements of the relationships of the metazoan hosts with their microenvironments including its microbiota and thus, would contribute greatly to the health state of men and animals.

References

  1. 1. JDAdams JrGarcia. C.2005Spirit, Mind and Body in Chumash Healing. Evid. based. complement. alternat. med. 2459463
  2. 2. YangY.2009Chinese Herbal Medicines. Comparisons and Characteristics. 2nd Editions. China. Churchill Livingstone Elsevier, pp
  3. 3. FornaroM.ClementiN.FornaroP.2009Medicine and Psychiatry in Western Culture: Ancient Greek Myths and Modern PrejudicesAnn. gen. psychiatry 8: 21.
  4. 4. YapijakisC.2009Hippocrates of Kos, the Father of Clinical Medicine, and Asclepiades of Bithynia, the Father of Molecular Medicine. In vivo507514
  5. 5. KellyK.The History of Medicine. Medicine Becomes a Science: 18401999 1999. Factson File Inc.
  6. 6. BernardC.1865Introduction à l’étude de la médecineexpérimentale. Available: http://classiques.uqac.ca/classiques/bernard_claude/intro_etude_medecine_exp/intro_medecine_exper.pdf
  7. 7. SelyeH.1936A Syndrome Produced by Diverse Nocuous AgentsNature32 EOF32 EOF
  8. 8. Vander Sherman and Luciano’s Human Physiology: The Mechanisms of Body Function.th Edition. The McGraw-Hill Companies, 728732
  9. 9. Chrousos GP, Gold PW1992The Concepts of Stress and Stress System Disorders. Overview of Physical and Behavioral Homeostasis.JAMA. 26712441252
  10. 10. CharmandariE.TsigosC.ChrousosG.2005Endocrinology of the Stress Response.Annu. rev. physiol. 6725984
  11. 11. Chrousos GP2009Stress and Disorders of Stress Systems. Nature rev. endoc. 5374381
  12. 12. MayerE. A.BDNaliboffChang. L.CoutinhoS. V.2001Stress and Irritable Bowel SyndromeAm. j. physiol. gastrointest. liver physiol. 280: G519G524.
  13. 13. GoymannW.WingfieldJ. C.2004Allostatic Load, Social Status and Stress Hormones: The Costs of Social Status MatterAnim. beh. 67591602
  14. 14. McEwen BS2000Allostasis, Allostatic Load, and the Aging Nervous System: Role of Excitatory Amino Acids and Excitotoxicity. Neurochem. res. 2512191231
  15. 15. Mayer EA2000The Neurobiology of Stress and Gastrointestinal DiseaseGut. 47861869
  16. 16. PapadimitriouA.PriftisK. N.2009Regulation of the Hypothalamic-Pituitary-Adrenal AxisNeuroimmunomodulation16265271
  17. 17. BhatiaV.TandonR. K.2005Stress and the Gastrointestinal Tract. J. gastroenterol. hepatol. 20332339
  18. 18. LaraucheM.KiankC.TacheY.2009Corticotropin Releasing Factor Signaling in Colon and Ileum: Regulation by Stress and Pathophysiological Implications.J. physiol. pharmacol. 60 (Suppl 7): 33 EOF46 EOF
  19. 19. LyteM.VulchanovaL.BrownD. R.2011Stress at the Intestinal Surface: Catecholamines and Mucosa-Bacteria Interactions.Cell tissue res. 3432332
  20. 20. Söderholm JD, Perdue MH2001Stress and Gastrointestinal Tract. II. Stress and Intestinal Barrier Function.Am. j. physiol. gastrointest. liver physiol. 280: G7G13.
  21. 21. DaviesE.MacKenzie. S. M.2003Extra-Adrenal Production of CorticosteroidsClin. Exp. pharmacol. physiol. 30437445
  22. 22. NotiM.SidlerD.BrunnerT.2009Extra-Adrenal Glucocorticoid Synthesis in the Intestinal Epithelium: More than a Drop in the Ocean? Semin. immunopathol. 31237248
  23. 23. AshidaH.OgawaM.KimM.MimuroH.SasakawaC.2011Bacteria and Host Interactions in the Gut Epithelial Barrier.Nat. chem. biol. 83645
  24. 24. SekirovI.RussellS. L.AntunesL. C.FinlayB. B.2010Gut Microbiota in Health and DiseasePhysiol. rev. 90859904
  25. 25. SudoN.ChidaY.AibaY.SonodaJ.OyamaN.YuX. N.KuboC.KogaY.2004Postnatal Microbial Colonization Programs the Hypothalamic-Pituitary-Adrenal System for Stress Response in Mice.J. physiol. 558263275
  26. 26. Bailey MT, Coe CL1999Maternal Separation Disrupts the Integrity of the Intestinal Microflora in Infant Rhesus Monkeys.Dev psychobiol. 35146155
  27. 27. FreestoneP. P.HaighR. D.WilliamsP. H.LyteM.1999Stimulation of Bacterial Growth by Heat-Stable Norepinophrine-Induced Autoinducers. FEMS microbial. letters. 1725360
  28. 28. Meddings JB, Swain MG2000Environmental Stress-Induced Gastrointestinal Permeability is Mediated by Endogenous Glucocorticoids in the RatGastroenterology11910191028
  29. 29. SpitzJ.HechtG.TaverasM.AoysE.AlverdyJ.1994The Effect of Dexamethasone Administration on Rat Intestinal Permeability: The Role of Bacterial Adherence.Gastroenterology1063541
  30. 30. SpitzJ. C.GhandiS.TaverasM.AoysE.AlverdyJ. C.1996Characteristics of the Intestinal Epithelial Barrier During Dietary Manipulation and Glucocorticoid Stress.Crit. care med. 24635641
  31. 31. EstienneM.ClaustreJ.Clain-GardechauxG.PaquetA.TachéY.FioramontiJ.PlaisanciéP.2010Maternal Deprivation Alters Epithelial Secretory Cell Lineages in Rat Duodenum: Role of CRF-Related PeptidesGut59744751
  32. 32. TachéY.MartinezV.MillionM.WangL.2001Stress and the Gastrointestinal Tract III. Stress-Related Alterations of Gut Motor Function: Role of Brain Corticotropin-Releasing Factor Receptors.Am. j. physiol. gastrointest. liver physiol. 280: G173G177.
  33. 33. TsukadaF.SugawaraM.KohnoH.OhkuboY.2001Evaluation of the Effects of Restraint and Footshock Stress on Small Intestinal Motility by an Improved Method Using a Radionuclide, 51 Cr, in the RatBiol. pharm. bull. 2448890
  34. 34. BradesiS.SchwetzI.EnnesH. S.LamyC. M.OhningG.FanselowM.PothoulakisC.Mc RobertsJ. A.MayerE. A.2005Repeated Exposure to Water Avoidance Stress in Rats: A New Model for Sustained Visceral HyperalgesiaAm. j. physiol. gastrointest. liver physiol. 289: G42G53.
  35. 35. Williams CL, Villar RG, Peterson JM, Burks TF1988Stress-Induced Changes in Intestinal Transit in the Rat: A Model for Iirritable Bowel Syndrome. Gastroenterology. 94611621
  36. 36. SaundersP. R.KoseckaU.Mc KayD. M.PerdueM. H.1994Acute Stressors Stimulate Ion Secretion and Increase Epithelial Permeability in Rat Intestine.Am. j. physiol. 267: G794G799.
  37. 37. MillionM.TachéY.AntonP.1999Susceptibility of Lewis and Fischer Rats to Stress-Induced Worsening of TNB-Colitis: Protective Role of Brain CRF.Am. j. physiol. 276: G1027G1036.
  38. 38. JDSöderholmStreutker. C.YangP. C.PatersonC.SinghP. K.Mc KayD. M.ShermanP. M.CroitoruK.PerdueM. H.2004Increased Epithelial Uptake of Protein Antigens in the İleum of Crohn’s Disease Mediated by Tumour Necrosis Factor Alpha. Gut. 5318171824
  39. 39. VicarioM.GuilarteM.AlonsoC.YangP. C.MartínezC.RamosL.LoboB.GonzálezA.GuilàM.PigrauM.SaperasE.AzpirozF.SantosJ.2010Chronological Assessment of Mast Cell-Mediated Gut Dysfunction and Mucosal Inflammation in a Rat Model of Chronic Psychosocial Stress. Brain behav. immun. 2411661175
  40. 40. VicarioM.AlonsoC.GuilarteM.SerraJ.MartínezC.González-CastroA. M.LoboB.AntolínM.AndreuA. L.García-ArumíE.CasellasM.SaperasE.MalageladaJ. R.AzpirozF.SantosJ.2012Chronic Psychosocial Stress Induces Reversible Mitochondrial Damage and Corticotropin-Releasing Factor Receptor Type-1 Upregulation in the Rat Intestine and IBS-like Gut Dysfunction. Psychoneuroendocrinology 376577
  41. 41. JDSöderholmYates. D. A.GareauM. G.YangP. C.MacQueen. G.PerdueM. H.2002Neonatal Maternal Separation Predisposes Adult Rats to Colonic Barrier Dysfunction in Response to Mild Stress.Am. j. physiol. gastrointest. liver physiol. 283: G1257G1263.
  42. 42. JDSöderholmYang. P. C.CeponisP.VohraA.RiddellR.ShermanP. M.PerdueM. H.2002Chronic Stress Induces Mast Cell-Dependent Bacterial Adherence and Initiates Mucosal Inflammation in Rat IntestineGastroenterology12310991108
  43. 43. GareauM. G.JuryJ.YangP. C.MacQueen. G.PerdueM. H.2006Neonatal Maternal Separation Causes Colonic Dysfunction in Rat Pups Including Impaired Host Resistance.Pediatr. res. 598388
  44. 44. GareauM. G.JuryJ.PerdueM. H.2007Neonatal Maternal Separation of Rat Pups Results in Abnormal Cholinergic Regulation of Epithelial PermeabilityAm. j. physiol. gastrointest. liver physiol. 293: G198G203.
  45. 45. IwasakiA.KelsallB. L.1999Mucosal Dendritic Cells: Their Specialized Role in Initiating T Cell Responses. Am. j. physiol. gastroenterol. liverphysiol. 276: G1074G1078.
  46. 46. Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB.1999Myofibroblasts. I. Paracrine Cells Important in Health and Disease. Am j. physiol. 277: C1C9.
  47. 47. Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB.1999Myofibroblasts. II. Intestinal Subepithelial Myofibroblasts. Am. j. physiol. 277: C183C201.
  48. 48. Stagg AJ, Hart AL, Knight SC, Kamm MA2003The Dendritic Cell: Its Role in Intestinal Inflammation and Relationship with Gut BacteriaGut. 5215221529
  49. 49. Catron DM, Itano AA, Pape KA, Mueller DL, Jenkins MK2004Visualizing the First 50 Hr of the Primary Immune Response to a Soluble Antigen.Immunity21341347
  50. 50. MacphersonA. J.UhrT.2004Induction of Protective IgA by Intestinal Dendritic Cells Carrying Commensal Bacteria.Science. 30316621665
  51. 51. SaadaJ. I.CABarreraReyes. V. E.AdegboyegaP. A.SuarezG.TamerisaR. A.PangK. F.BlandD. A.MifflinR. C. D. I.MariJ. F.PowellD. W.2004Intestinal Myofibroblasts and Immune Tolerance. Ann. n. y. acad sci. 1029379381
  52. 52. Niess JH, Reinecker HC2005Lamina Propria Dendritic Cells in the Physiology and Pathology of the Gastrointestinal Tract.Curr. opin. gastroenterol. 21687691
  53. 53. LeonF.SymythiesL. E.SmithP. D.KelsallB. L.2006Involvement of Dendritic Cells in the Pathogenesis of Inflammatory Bowel DiseasesAdv. exp. med. biol. 579117132
  54. 54. InmanC. F.SinghaLewis. M.BradleyB.StokesC.BaileyM.2010Dendritic Cells Interact with CD4 T Cells in Intestinal MucosaJ. leukocyte biol. 88571578
  55. 55. ManicassamyS.ReizisB.RavindranR.NakayaH.Salazar-GonzalezR. M.WangY. C.PulendranB.2010Activation of Beta-Catenin in Dendritic Cells Regulates Immunity Versus Tolerance in the Intestine.Science. 329849853
  56. 56. Perdue MH1999Mucosal Immunity and Inflammation III. The Mucosal Antigen Barrier: Cross Talk with Mucosal Cytokines Am. j. physiol. gastrointest. liver physiol. 277: G1G5.
  57. 57. Mason KL, Huffnagle GB, Noverr MC, Kao JY2008Overview of Gut ImmunologyIn: Huffnagle GB, Noverr MC, editors. GI Microbiota and Regulation of the Immune System: Advances in Experimental Medicine and Biology635Landes Bioscience Springer Science+Business Media. 110
  58. 58. BoudryG.JuryJ.YangP. C.PerdueM. H.2007Chronic Psychological Stress Alters Epithelial Cell Turn-Over in Rat Ileum.Am. j. physiol. gastrointest. liver physiol. 292: G1228G1232.
  59. 59. Cameron HL, Perdue MH2005Stress Impairs Murine İntestinal Barrier Function: Improvement by Glucagon-Like Peptide-2. J. pharmacol. exp. ther. 314214220
  60. 60. Kiliaan AJ, Saunders PR, Bijlsma PB, Berin MC, Taminiau JA, Groot JA, Perdue MH.1998Stress Stimulates Transepithelial Macromolecular Uptake in Rat Jejunum.Am. j. physiol. gastrointest. liver. physiol. 275: G1037G1044.
  61. 61. SantosJ.BenjaminM.YangP. C.PriorT.PerdueM. H.2000Chronic Stress Impairs Rat Growth and Jejunal Epithelial Barrier Function: Role of Mast Cells.Am. j. physiol. gastrointest. liver physiol. 278: G847G854.
  62. 62. Hung CR.1998Low Susceptibility of Stress Ulcer in Diabetic Rats: Role of Cholinergic Gastric MotilityChin. j. physiol. 41151159
  63. 63. BabygirijaR.ZhengJ.BülbülM.LudwigK.TakahashiT.2010Beneficial Effects of Social Attachment to Overcome Daily StressBrain res.13524349
  64. 64. ZhengJ.BabygirijaR.BülbülM.CerjakD.LudwigK.TakahashiT.2010Hypothalamic Oxytocin Mediates Adaptation Mechanism Against Chronic Stress in RatsAm. j. physiol. gastrointest. liver physiol. 299: G946G953.
  65. 65. WallonC.JDSöderholm2009Corticotropin-Releasing Hormone and Mast Cells in the Regulation of Mucosal Barrier Function in the Human ColonAnn. n. y. acad. sci. 1165206210
  66. 66. SuzukiK.HarasawaR.YoshitakeY.MitsuokaT.1983Effects of Crowding and Heat Stress on Intestinal flora, Body Weight Gain, and Feed Efficiency of Growing Rats and Chicks.Jpn. j. vet. sci. 45331338
  67. 67. Tannock GW1997Modification of the Normal Microbiota by Diet, Stress, Antimicrobial Agents, and Probiotics. In: Mackie RI, White BA, Isaacson RE, editors. Gastrointestinal Microbiology. New York. Chapman & Hall, 434466
  68. 68. Bailey MT, Lubach GR, Coe CL2004Prenatal Stress Alters Bacterial Colonization of the Gut in Infant Monkeys.J. pediatr. gastroenterol. nutr. 38414421
  69. 69. LutgendorffF.AkkermansL. M.JDSöderholm2008The Role of Microbiota and Probiotics in Stress-Induced Gastro-Intestinal Damage.Curr. mol. med. 84282298
  70. 70. MazzonE.SturnioloG. C.PuzzoloD.FrisinaN.FriesW.2002Effect of Stress on the Paracellular Barrier in the Rat IleumGut. 51507513
  71. 71. WangF.GrahamW. V.WangY.WitkowskiE. D.SchwarzB. T.TurnerJ. R.2005Interferon-Gamma and Tumor Necrosis Factor-Alpha Synergize to Induce Intestinal Epithelial Barrier Dysfunction by Up-regulating Myosin Light Chain Kinase Expression.Am. j.pathol. 166409419
  72. 72. GrahamW. V.WangF.ClayburghD. R.ChengJ. X.YoonB.WangY.LinA.TurnerJ. R.2006Tumor Necrosis Factor-Induced Long Myosin Light Chain Kinase Transcription is Regulated by Differentiation-Dependent Signaling Events. Characterization of the Human Long Myosin Light Chain Kinase Promoter.J. biol. chem. 2812620526215
  73. 73. Al-SadiR.YeD.DokladnyK.MaT. Y.2008Mechanism of IL-1 Beta-Induced Increase in Intestinal Epithelial Tight Junction Permeability.J. immunol. 18056535661
  74. 74. Ait-BelgnaouiA.BradesiS.FioramontiJ.TheodorouV.BuenoL.2005Acute Stress-Induced Hypersensitivity to Colonic Distension Depends upon Increase in Paracellular Permeability: Role of Myosin Light Chain Kinase.Pain113141147
  75. 75. MazzonE.CuzzocreaS.2008Role of TNF-Alpha in Ileum Tight Junction Alteration in Mouse Model of Restraint Stress.Am. j. physiol. gastrointest. liver physiol. 294: G1268G1280.
  76. 76. MatsuoK.ZhangX.OnoY.NagatomiR.2009Acute Stress-Induced Colonic Tissue HSP70 Expression Requires Commensal Bacterial Components and Intrinsic GlucocorticoidBrain behav. immune. 23108115
  77. 77. MABoivinYe. D.KennedyJ. C.Al-SadiR.ShepelaC.MaT. Y.2007Mechanism of Glucocorticoid Regulation of the Intestinal Tight Junction BarrierAm. j physiol. gastrointest. liver physiol. 292: G590G598.
  78. 78. Barclay GR, Turnberg LA1987Effect of Psychological Stress on Salt and Water Transport in the Human Jejunum.Gastroenterology939197
  79. 79. Barclay GR, Turnberg LA1988Effect of Cold-Induced Pain on Salt and Water Transport in the Human Jejunum.Gastroenterology94994998
  80. 80. SantosJ.SaundersP. R.HanssenN. P.YangP. C.YatesD.GrootJ. A.PerdueM. H.1999Corticotropin-Releasing Hormone Mimics Stress-Induced Colonic Epithelial Pathophysiology in the Rat Am. j. physiol. 277: G391G399.
  81. 81. SaundersP. R.SantosJ.HanssenN. P.YatesD.GrootJ. A.PerdueM. H.2002Physical and Psychological Stress in Rats Enhances Colonic Epithelial Permeability via Peripheral CRH.Dig. dis. sci. 47208215
  82. 82. LaraucheM.GourcerolG.WangL.PambukchianK.BrunnhuberS.AdelsonD. W.RivierJ.MillionM.TachéY.2009A Cortagine CRF1 Agonist, Induces Stresslike Alterations of Colonic Function and Visceral Hypersensitivity in Rodents Primarily Through Peripheral Pathways. Am. j. physiol. gastrointest. liver physiol. 297: G215G227.
  83. 83. CharneyA. N.MDKinseyMyers. L.GainnellaR. A.GotsR. E.1975Na+-K+-Activated Adenosine Triphosphatase and Intestinal Electrolyte Transport. Effect of Adrenal Steroids. J. clin invest. 56653660
  84. 84. MarnaneW. G.TaiY. H.DeckerR. A.BoedekerE. C.CharneyA. N.DonowitzM.1981Methylprednisolone Stimulation of Guanylate Cyclase Activity in Rat Small Intestinal Mucosa: Possible Role in Electrolyte Transport.Gastroenterology8190100
  85. 85. TaiY. H.DeckerR. A.MarnaneW. G.CharneyA. N.DonowitzM.1981Effects of Methylprednisolone on Electrolyte Transport by In Vitro Rat Ileum.Am. j. physiol. 240: G365G370.
  86. 86. YatesD. A.SantosJ.JDSöderholmPerdue. M. H.2001Adaptation of stress-induced mucosal pathophysiology in rat colon involves opioid pathwaysAm. j. physiol. gastrointest. liver physiol. 281: G124G128.
  87. 87. SantosJ.SaperasE.NogueirasC.MourelleM.Antoli´nM.CadahiaA.MalageladaJ. R.1998Release of Mast Cell Mediators into the Jejunum by Cold Pain Stress in Humans.Gastroenterology114640648
  88. 88. Gerova VA, Stoynov SG, Katsarov DS, Svinarov DA2011Increased Intestinal Permeability in Inflammatory Bowel Diseases Assessed by Iohexol TestWorld j. gastroenterol. 1722112215
  89. 89. GecseK.RókaR.SéraT.RosztóczyA.AnnaháziA.IzbékiF.NagyF.MolnárT.SzepesZ.PávicsL.BuenoL.WittmannT.2012Leaky Gut in Patients with Diarrhea-Predominant Irritable Bowel Syndrome and Inactive Ulcerative Colitis. Digestion. 854046
  90. 90. BagchiD.CarrylO. R.TranM. X.BagchiM.GargA.MMMilnesWilliams. C. B.BalmooriJ.BagchiD. J.MitraS.StohsS. J.1999Acute and Chronic Stress-Induced Oxidative Gastrointestinal Mucosal Injury in Rats and Protection by Bismuth Subsalicylate. Mol. cell biochem. 196109116
  91. 91. BarreauF.FerrierL.FioramontiJ.BuenoL.2004Neonatal Maternal Deprivation Triggers Long Term Alterations in Colonic Epithelial Barrier and Mucosal Immunity in RatsGut. 53501506
  92. 92. ZhangZ. W.LvZ. H.LiJ. L.LiS.XuS. W.WangX. L.2011Effects of Cold Stress on Nitric Oxide in Duodenum of ChicksPoult. sci. 9015551561
  93. 93. WitthöftT.EckmannL.KimJ. M.KagnoffM. F.1998Enteroinvasive Bacteria Directly Activate Expression of INOS and NO Production in Human Colon Epithelial Cells.Am. j. physiol. 275: G564G571.
  94. 94. SantosJ.YangP. C.JDSoderholmBenjamin. M.PerdueM. H.2001Role of Mast Cells in Chronic Stress Induced Colonic Epithelial Barrier Dysfunction in the RatGut 48630636
  95. 95. Perdue MH, McKay DM1994Integrative Immunophysiology in the Intestinal Mucosa.Am. j. physiol. 267: G151G165.
  96. 96. CastagliuoloI.WershilB. K.KaralisK.PashaA.NikulassonS. T.PothoulakisC.1998Colonic Mucin Release in Response to Immobilization Stress is Mast Cell DependentAm. j. physiol. gastrointest. liver physiol. 274: G1094G1100.
  97. 97. Kim DH, Cho YJ, Kim JH, Kim YB, Lee KJ2010Stress-Induced Alterations in Mast Cell Numbers and Proteinase-Activated Receptor-2 Expression of the Colon: Role of Corticotrophin-Releasing Factor.J. korean med. sci. 2513301335
  98. 98. RókaR.Ait-BelgnaouiA.Salvador-CartierC.Garcia-VillarR.FioramontiJ.EutamèneH.BuenoL.2007Dexamethasone Prevents Visceral Hyperalgesia but not Colonic Permeability Increase Induced by Luminal Protease-Activated Receptor-2 Agonist in Rats. Gut. 5610721078
  99. 99. Wilson LM, Baldwin AL1999Environmental Stress Causes Mast Cell Degranulation, Endothelial and Epithelial Changes, and Edema in the Rat Intestinal MucosaMicrocirculation6189198
  100. 100. JorgeE.FernándezJ. A.TorresR.VergaraP.MartinM. T.2010Functional Changes Induced by Psychological Stress are not Enough to Cause Intestinal Inflammation in Sprague-Dawley RatsNeurogastroenterol. motil. 22: e241e250.
  101. 101. GodotV.GarciaG.CapelF.ArockM.Durant-GasselinI.Asselin-LabatM. L.EmilieD.HumbertM.2006Dexamethasone and IL-10 Stimulate Glucocorticoid-Induced Leucine Zipper Synthesis by Human Mast Cells. Allergy 61886890
  102. 102. RijnierseA.ASKosterNijkamp. F. P.KraneveldA. D.2006TNF-Alpha is Crucial for the Development of Mast Cell-Dependent Colitis in Mice.Am. j. physiol. gastrointest. liver physiol. 291: G969G976.
  103. 103. EutameneH.TheodorouV.FioramontiJ.BuenoL.2003Acute Stress Modulates the Histamine Content of Mast Cells in the Gastrointestinal Tract Through Interleukin-1 and Corticotropin-Releasing Factor Release in Rats.J. physiol. 553959966
  104. 104. WallonC.YangP. C.KeitaA. V.EricsonA. C.Mc KayD. M.ShermanP. M.PerdueM. H.JDSöderholm2008Corticotropin-Releasing Hormone (CRH) Regulates Macromolecular Permeability via Mast Cells in Normal Human Colonic Biopsies In VitroGut575058
  105. 105. MaloyK. J.PowrieF.2011Intestinal Homeostasis and Its Breakdown in Inflammatory Bowel Disease.Nature474298306
  106. 106. MAMc GuckinLindén. S. K.SuttonP.FlorinT. H.2011Mucin Dynamics and Enteric PathogensNat. rev microbiol. 9265278
  107. 107. DharmaniP.SrivastavaV.Kissoon-SinghV.ChadeeK.2009Role of Intestinal Mucins in Innate Host Defense Mechanisms Against PathogensJ. innate immun. 1123135
  108. 108. CastagliuoloI.LamontJ. T.QiuB.FlemingS. M.BhaskarK. R.NikulassonS. T.KornetskyC.PothoulakisC.1996Acute Stress Causes Mucin Release From Rat Colon: Role of Corticotropin Releasing Factor and Mast Cells.Am. j. phyiol. 271: G884G892.
  109. 109. O’MalleyD.Julio-PieperM.GibneyS. M.DinanT. G.CryanJ. F.2010Distinct Alterations in Colonic Morphology and Physiology in Two Rat Models of Enhanced Stress-Induced Anxiety and Depression-like BehaviourStress132114122
  110. 110. Finnie IA, Campbell BJ, Taylor BA, Milton JD, Sadek SK, Yu LG, Rhodes JM1996Stimulation of Colonic Mucin Synthesis by Corticosteroids and Nicotine.Clin. sci. (Lond) 91359364
  111. 111. TsukamotoK.NakadeY.MantyhC.LudwigK.PappasT. N.TakahashiT.2006Peripherally Administered CRF Stimulates Colonic Motility via Central CRF Receptors and Vagal Pathways in Conscious RatsAm. j. physiol. regul. integr. comp. physiol. 290: R1537R1541.
  112. 112. MichelsenK. S.ArditiM.2007Toll-like Receptors and Innate Immunity in Gut Homeostasis and Pathology.Curr. opin. hematol. 144854
  113. 113. GrenhamS.ClarkeG.CryanJ. F.DinanT. G.2011Brain-Gut-Microbe Communication in Health and Disease.Front. physiol. 2114
  114. 114. ShiboletO.PodolskyD. K.2007TLRs in the Gut. IV. Negative Regulation of Toll-like Receptors and Intestinal Homeostasis: Addition by Subtraction.Am. j. physiol. gastrointest. liver physiol. 292: G1469G1473.
  115. 115. Jarillo-LunaA.Rivera-AguilarV.Martìnez-CarrilloB. E.Barbosa-CabreraE.GarfiasH. R.Campos-RodríguezR.2008Effect of Restraint Stress on the Population of Intestinal Intraepithelial Lymphocytes in Mice. Brain behav. immun. 22265275
  116. 116. Mc KernanD. P.NolanA.BrintE. K.O’MahonyS. M.HylandN. P.CryanJ. F.DinanT. G.2009Toll-Like Receptor mRNA Expression is Selectively Increased in the Colonic Mucosa of Two Animal Models Relevant to Irritable Bowel Syndrome.PLoS ONEe8226 EOFAvailable: http://www.plosone.org/article/info%3adoi%2fF10.1371%2Fjournal.pone.0008226.
  117. 117. ZhangY.WoodruffM.ZhangY.MiaoJ.HanleyG.StuartC.ZengX.SprabhajarS.MoormanJ.ZhaoB.YinD.2008Toll-like Receptor 4 Mediates Chronic Restraint Stress-Induced Immune Suppression. J. neuroimmunol. 194115122
  118. 118. BaileyM. T.EnglerH.PowellN. D.PadgettD. A.SheridanJ. F.2007Repeated Social Defeat Increases the Bactericidal Activity of Splenic Macrophages Through a Toll-like Receptor-Dependent Pathway.Am. j. physiol. regul. integr. comp. physiol. 293: R1180R1190.
  119. 119. Powell ND, Bailey MT, Mays JW, Stiner-Jones LM, Hanke ML, Padgett DA, Sheridan JF2009Repeated Social Defeat Activates Dendritic Cells and Enhances Toll-like Receptor Dependent Cytokine SecretionBrain behav. immun. 23225231
  120. 120. Mc KernanD. P.GasznerG.QuigleyE. M.CryanJ. F.DinanT. G.2011Altered Peripheral Toll-like Receptor Responses in the Irritable Bowel Syndrome.Aliment. pharmacol. ther. 3310451052
  121. 121. YangP. C.JuryJ.JDSöderholmSherman. P. M.MckayD. M.PerdueM. H.2006Chronic Psychological Stress in Rats Induces Intestinal Sensitization to Luminal AntigensAm. j. pathol. 168104114
  122. 122. VelinA. K.EricsonA. C.BraafY.WallonC.JDSöderholm2004Increased Antigen and Bacterial Uptake in Follicle Associated Epithelium Induced by Chronic Psychological Stress in RatsGut. 53494500
  123. 123. KeitaA. V.SalimS. Y.JiangT.YangP. C.FranzénL.SöderkvistP.MagnussonK. E.JDSöderholm2008Increased Uptake of Non-pathogenic E. Coli via the Follicle-Associated Epithelium in Longstanding Ileal Crohn’s DiseaseJ. pathol. 215135144
  124. 124. Keita AV, Söderholm JD, Ericson AC2010Stress-Induced Barrier Disruption of Rat Follicle-Associated Epithelium Involves Corticotropin-Releasing Hormone, Acetylcholine, Substance P, and Mast CellsNeurogastroenterol. motil. 22770778
  125. 125. ZhengP. Y.BSFengOluwole. C.StruiksmaS.ChenX.LiP.TangS. G.YangP. C.2009Psychological Stress Induces Eosinophils to Produce Corticotrophin Releasing Hormone in the Intestine. Gut. 5814731479
  126. 126. KoslowskiM. J.BeisnerJ.StangeE. F.WehkampJ.2010Innate Antimicrobial Host Defense in Small Intestinal Crohn’s DiseaseInt. j. med. microbiol. 3003440
  127. 127. Cash HL, Whitham CV, Behrendt CL, Hooper LV2006Symbiotic Bacteria Direct Expression of an Intestinal Bactericidal Lectin.Science. 31311261130
  128. 128. AyabeT.SatchellD. P.WilsonC. L.ParksW. C.MESelstedOuellette. A. J.2000Secretion of Microbicidal Alpha-Defensins by Intestinal Paneth Cells in Response to Bacteria.Nat. immuno1. 199100
  129. 129. MacphersonA. J.SlackE.2007The Functional Interactions of Commensal Bacteria with Intestinal Secretory IgA.Curr. opin. gastroenterol. 236738
  130. 130. KellyP.FeakinsR.DomizioP.MurphyJ.BevinsC.WilsonJ.Mc PhailP.PoulsomR.DhaliwalW.2004Paneth Cell Granule Depletion in Human Small Intestine under Infective and Nutritional Stress. Clin. exp. immunol. 135303309
  131. 131. AlonsoC.GuilarteM.VicarioM.RamosL.RamadanZ.AntolínM.MartínezC.RezziS.SaperasE.KochharS.SantosJ.MalageladaJ. R.2008Maladaptive Intestinal Epithelial Responses to Life Stress may Predispose Healthy Women to Gut Mucosal Inflammation. Gastroenterology. 135163172
  132. 132. MurosakiS.Inagaki-OharaK.KusakaH.IkedaH.YoshikaiY.1997Apoptosis of Intestinal Intraepithelial Lymphocytes Induced by Exogenous and Endogenous Glucocorticoids.Microbiol. immunol. 41139148
  133. 133. FukuzukaK.EdwardsC. K.3rd Clare-SalzerM.CopelandE. M.3rd MoldawerL. L.MozingoD. W.2000Glucocorticoid and Fas Ligand Induced Mucosal Lymphocyte Apoptosis After Burn Injury.J. trauma. 49710716
  134. 134. ReberS. O.PetersS.SlatteryD. A.HofmannC.SchölmerichJ.NeumannI. D.ObermeierF.2011Mucosal Immunosuppression and Epithelial Barrier Defects are Key Events in Murine Psychosocial Stress-Induced ColitisBrain behav. immun. 2511531161
  135. 135. MotykaB.BhogalH. S.JDReynolds1995Apoptosis of Ileal Peyer’s Patch B Cells is Increased by Glucocorticoids or Anti-immunoglobulin Antibodies.Eur. j. immunol. 2518651871
  136. 136. Ruiz-SantanaS.LopezA.TorresS.ReyA.LosadaA.LatasaL.ManzanoJ. L.Diaz-ChicoB. N.2001Prevention of Dexamethasone Induced Lymphocytic Apoptosis in the Intestine and in Peyer Patches by Enteral Nutrition.J. parenter. enteral nutr. 25338345
  137. 137. VaughnJ.1961Experimental Eosinophilia: Local Tissue Reactions to Ascaris ExtractsJ. allergy. 32501513
  138. 138. BrowaeysJ.WallonD.1958Éosinophilies tissulaires du rat a l’état normal et dans les éosinopénies sanguine. Le sang 29686695
  139. 139. Elftman MD, Norbury CC, Bonneau RH, Truckenmiller ME2007Corticosterone Impairs Dendritic Cell Maturation and FunctionImmunology. 122279290
  140. 140. BSMc EwenBiron.CABrunsonK. W.BullochK.ChambersW. H.DhabharF. S.GoldfarbR. H.KitsonR. P.MillerA. H.SpencerR. L.WeissJ. M.1997The Role of Adrenocorticoids as Modulators of Immune Function in Health and Disease: Neural, Endocrine and Immune Interactions.Brain res. rev. 2379133
  141. 141. ToftP.LillevangS. T.TønnesenE.SvendsenP.HöhndorfK.1993Redistribution of Lymphocytes Following E. Coli Sepsis. Scand. j. immunol. 38541545
  142. 142. ToftP.SvendsenP.TønnesenE.RasmussenJ. W.ChristensenN. J.1993Redistribution of Lymphocytes after Major Surgical Stress. Acta anaesthesiol. scand. 37245249
  143. 143. Tlaskalová-HogenováH.StepánkováR.HudcovicT.TuckováL.CukrowskaB.Lodinová-ZádníkováR.KozákováH.RossmannP.BártováJ.SokolD.FundaD. P.BorovskáD.RehákováZ.SinkoraJ.HofmanJ.DrastichP.KokesováA.2004Commensal Bacteria (Normal Microflora), Mucosal Immunity and Chronic Inflammatory and Autoimmune Diseases. Immunol. lett. 9397108
  144. 144. EckburgP. B.BikE. M.BernsteinC. N.PurdomE.DethlefsenL.SergentM.GillS. R.NelsonK. E.RelmanD. A.2005Diversity of Human Intestinal Microbial Flora. Science. 30816351638
  145. 145. Ley RE, Peterson DA, Gordon JI2006Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human IntestineCell124837848
  146. 146. O’Hara AM, Shanahan EM2006The gut flora as a forgotten organEMBO rep. 7688693
  147. 147. HolzapfelW. H.HabererP.SnelJ.SchillingerU.JosH. J.Huisin’t.Veld1998Overview of Gut Flora and Probiotics. Int. j. food microbiol. 4185101
  148. 148. MackieR. I.SghirA.GaskinsH. R.1999Developmental Microbial Ecology of the Neonatal Gastrointestinal Tract.Am. j. clin. nutr. 69(suppl.): 10351045
  149. 149. GuerreroR.BerlengaM.2006Life’s Unity and Flexibility: The Ecological Link. Int. microbiol. 9225235
  150. 150. Savage DC1999Mucosal Microbiota. In: Ogra PL, Mestecky J, Lamm ME, Strober W, Bienenstock J, McGhee J, editors. Mucosal immunology. New York: Academic Press. 1930pp.
  151. 151. GillS. R.PopM.DeboyR. T.EckburgP. B.TurnbaughP. J.BSSamuelGordon. J. I.RelmanD. A.Fraser-LiggettC. M.NelsonK. E.2006Meta Genomic Analysis of the Human Distal Gut Microbiome. Science. 31213551359
  152. 152. Hooper LV, Gordon JI2001Commensal Host-Bacterial Relationships in the Gut.Science. 29211151118
  153. 153. TurnbaughP. J.LeyR. E.HamadyM.Fraser-LiggettC. M.KnightR.GordonJ. I.2007The Human Microbiome Project. Nature. 449804810
  154. 154. Berg RD1996The Indigenous Gastrointestinal Microflora.Trends microbial. 4430435
  155. 155. MJClaessonCusack. S.O’SullivanO.Greene-DinizR.De WeerdH.FlanneryE.MarchesiR.FalushD.DinanT.FitzgeraldG.StantonC.Van SinderenD.O’ConnorM.HarnedyN.O’ConnorK.HenryC.O’MahonyD.FitzgeraldA. P.ShanahanF.TwomeyC.HillC.RossR. P.O’TooleP. W.2011Composition, Variability, and Temporal Stability of the Intestinal Microbiota of the Elderly. Proc. natl. acad. sci. 108(S1): 4586-4591.
  156. 156. PalmerC.BikE. M.Di GiulioD. B.RelmanD. A.BrownP. O.2007Development of the Human Infant Intestinal MicrobiotaPLoS Biol 5(7): e177. doi:journal.pbio.0050177.
  157. 157. AnderssonA. F.LinbergM.JakobssonH.BäkhedF.NyrénP.EngstrandL.2008Comparative Analysis of Human Gut Microbiota by Barcoded PyrosequencingPLoS One. 3(7): Article ID e2836.
  158. 158. Frank DN, Pace NR2008Gastrointestinal Microbiology Enters the Metagenomics EraCurr. opin. gastroent. 24410
  159. 159. Miller TL, Wolin MJ1983Stability of Methanobrevibacter Smithii Populations in the Microbial Flora Excreted from the Human Large BowelAppl. environ. microbiol. 45317318
  160. 160. Simon GL, Gorbach SL1984Intestinal Flora in Health and Disease.Gastroenterology86174193
  161. 161. DubosR.SchaedlerR. W.CostelloR.HoetP.1965Indigenous, Normal, and Autochthonous Flora of the Gastrointestinal Tract. J. exp. med. 1226776
  162. 162. KhouryK. A.FlochM. H.HershT.1969Small Intestinal Mucosal Cell Proliferation and Bacterial Flora in the Conventionalization of the Germfree MouseJ. exp. med. 130659670
  163. 163. GuarnerF.MalageladaJ. R.2003Gut Flora in Health and Disease.Lancet361512519
  164. 164. Wotsmann BS, Kardoss EB, Knight PL1968Cecal Enlargement, Cardiac Output and O2 Consumption in Germfree Rats. Proc. soc. exp. biol. med. 128137140
  165. 165. HooperL. V.MacphersonA. J.2010Immune Adaptations that Maintain Homeostasis with the Intestinal MicrobiotaNat. rev. immunol. 10159169
  166. 166. Gilmore MS, Ferretti JJ2003Microbiology. The Thin Line Between Gut Commensal and Pathogen. Science. 29919992002
  167. 167. Tannock GW2005New Perceptions of the Gut Microbiota: Implications for Future Research.Gastroenterol. clin. north am. 34361382
  168. 168. MartinF. P.SprengerN.MontoliuI.RezziS.KochharS.NicholsonJ. K.2010Dietary Modulation of Gut Functional Ecology Studied by Fecal MetabonomicsJ. proteome res. 952845295
  169. 169. HeathP.ClausS. P.2011Assessing hepatic metabolic changes during progressive colonization of germ-free mouse by 1H NMR spectroscopy.J. vis exp. (58) pii: 3642. doi:
  170. 170. ClausS. P.TsangT. M.WangY.CloarecO.SkordiE.MartinF. P.RezziS.RossA.KochharS.HolmesE.NicholsonJ. K.2008Systemic Multicompartmental Effects of the Gut Microbiome on Mouse Metabolic Phenotypes. Mol. syst. biol. 4: 219.
  171. 171. ZhengX.XieG.ZhaoA.ZhaoL.YaoC.ChiuN. H.ZhouZ.BaoY.JiaW.NicholsonJ. K.JiaW.2011The Footprints of Gut Microbial-Mammalian Co-metabolism. J. proteome res. 1055125522
  172. 172. Keeney KM, Finlay BB2011Enteric Pathogen Exploitation of the Microbiota-Generated Nutrient Environment of the GutCurr. opin. microbiol. 149298
  173. 173. BouskraD.BrezillonC.BerardM.WertsC.VaronaR.BonecaI. G.EberlG.2008Lymphoid Tissue Genesis Induced by Commensals Through NOD1 Regulates Intestinal HomeostasisNature456507510
  174. 174. Macpherson AJ, Harris NL2004Interactions Between Commensal Intestinal Bacteria and the Immune SystemNat. rev. immunol. 4478485
  175. 175. Vijay-KumarM.JDAitkenCarvalho. F. A.CullenderT. C.MwangiS.SrinivasanS.SitaramanS. V.KnightR.LeyR. E.GewirtzA. T.2010Metabolic Syndrome and Altered Gut Microbiota in Mice Lacking Toll-like Receptor 5. Science. 328228231
  176. 176. AtarashiK.NishimuraJ.ShimaT.UmesakiY.YamamotoM.OnoueM.YagitaH.IshiiN.EvansR.HondaK.et al.2008ATP Drives Lamina Propria T(H)17 Cell Differentiation. Nature. 455808812
  177. 177. ChowJ.MazmanianS. K.2009Getting the Bugs out of the Immune System: Do Bacterial Microbiota “Fix” Intestinal T Cell Responses? Cell host microbe. 5812
  178. 178. Fetissov and Déchelotte2011The New Link Between Gut-Brain Axis and Neuropsychiatric DisordersCurr. opin. clin. nutr. metab. care. 14477482
  179. 179. HooperL. V.WongM. H.ThelinA.HanssonL.FalkP. G.GordonJ. I.2001Molecular Analysis of Commensal Host-Microbial Relationships in the Intestine.Science 2881884
  180. 180. MozešS.BujňákováD.ŠefčikováZ.Kmet’V.2008Developmental Changes of Gut Microflora and Enzyme Activity in Rat Pups Exposed to Fat-Rich DietObesity1626102615
  181. 181. TurnbaughP. J.HamadyM.YatsunenkoT.CantarelB. L.DuncanA.LeyR. E.SoginM. L.JonesW. J.BARoeAffourtit. J. P.EgholmM.HenrissatB.HeathA. C.KnightR.GordonJ. I.2009A Core Gut Microbiome in Obese and Lean Twins. Nature. 457480484
  182. 182. ŠefčikováZ.KmeŤ. V.BujňákováD.RačekL’.MozešŠ.2010Development of Gut Microflora in Obese and Lean RatsFolia microbiol. 55373375
  183. 183. MancoM.PutignaniL.BottazzoG. F.2010Gut Microbiota, Lipopolysaccharides, and Innate Immunity in the Pathogenesis of Obesity and Cardiovascular Risk. Endocr. rev. 31817844
  184. 184. TurnbaughP.LeyR. E.MAMahowaldMagrini. V.MardisE. R.GordonJ. I.2006An Obesity-Associated Gut Microbiome with Increased Capacity for Energy Harvest.Nature44410271031
  185. 185. BäckhedF.DingH.WangT.HooperL. V.KohG. Y.NagyA.SemenkovichC. F.GordonJ. I.2004The Gut Microbiota as An Environmental Factor that Regulates Fat StorageProc. natl. acad. sci. 21571815723
  186. 186. PrinsA. .2011The Brain-Gut Interaction: The Conversation and the ImplicationsS. afr. j. clin. nutr. 24814
  187. 187. O’Mahony SM, Hyland NP, Dinan TG, Cryan JF.2011Maternal Separation as A Model of Brain-Gut Axis DysfunctionPsychopharmacology2147188
  188. 188. SudoN.2006Stress and Gut Microbiota: Does Postnatal Microbial Colonization Programs the Hypothalamic-Pituitary-Adrenal System for Stress Response? International Congress Series 1287350354
  189. 189. BravoJ. A.ForsytheP.ChewM. V.EscaravageE.SavignacH. M.DinanT. G.BienenstockJ.CryanJ. F.2011Ingestion of Lactobacillus Strain Regulates Emotional Behavior and Central GABA Receptor Expression in a Mouse Via the Vagus NerveProc. natl. acad. sci. 1081605016055
  190. 190. ForsytheP.SudoN.DinanT.TaylorV. H.BienenstockJ.2010Mood and Gut Feelings. Brain behav. immun. 241916
  191. 191. BaileyM. T.DowdS. E.ParryN. M.JDGalleySchauer. D. B.LyteM.2010Stressor Exposure Disrupts Commensal Microbial Populations in the Intestines and Leads to Increased Colonization by Citrobacter Rodentium.Infect. immun. 7815091519
  192. 192. Tannock GW, Savage DC1974Influences of Dietary and Environmental Stress on Microbial Populations in the Murine Gastrointestinal TractInfect, immun. 9591598
  193. 193. BıyıkH.BalkayaM.ÜnsalH.ÜnsalC.2005The Effects of Qualitative and Quantitative Protein Malnutrition on Cecal Microbiota in Wistar Rats with or without Neutrophil Suppression. Turk. j. vet. anim. sci. 29767773
  194. 194. ÜnsalH.BalkayaM.BıyıkH.ÜnsalC.BasbulbulG.PoyrazogluE.KozacıL. D.2009Time-dependent Effects of Dietary Qualitative and Quantitative Protein Malnutrition on Some Members of the Cecal Microbiota in Male Wistar Rats. Microb. ecol. health dis. 2144449
  195. 195. ÜnsalH.ÇöteliogluÜ.2007The Effects of Food Restriction on Some Biochemical Parameters and Certain Bacterial Groups in the Cecum in Sprague Dawley RatsMicrob. ecol. health dis. 191724
  196. 196. LyteM.ErnstS.1992Catecholamine Induced Growth of Gram Negative Bacteria.Life sci. 50302312
  197. 197. BaileyM. T.DowdS. E.JDGalleyHufnagle. A. R.AllenR. G.LyteM.2011Exposure to a Social Stressor Alters the Structure of the Intestinal Microbiota: Implications for Stressor-Induced ImmunomodulationBrain behave. immun. 25397407
  198. 198. O’MahonyS. M.MarchesiJ. R.ScullyP.CodlingC.CeolhoA. M.QuigleyE. M.CryanJ. F.DinanT. G.2009Early Life Stress Alters Behavior, Immunity, and Microbiota in Rats: Implications for Irritable Bowel Syndrome and Psychiatric IllnessesBiol. psychiatr. 65263267
  199. 199. Knowles SR, Nelson EA, Palombo EA2008Investigating the Role of Perceived Stress on Bacterial Flora Activity and Salivary Cortisol Secretion: A Possible Mechanism Underlying Susceptibility to IllnessBiol. psychol. 77132137
  200. 200. ÜnsalH.BalkayaM.ÜnsalC.BıyıkH.BaşbülbülG.PoyrazoğluE.2008The Short-term Effects of Different Doses of Dexamethasone on the Numbers of Some Bacteria in the Ileum. Dig. dis. sci. 5318421845
  201. 201. KirimliogluV.KirimliogluH.YilmazS.PiskinT.TekerekogluS.BayindirY.2006Effect of Steroid on Mitochondrial Oxidative Stress Enzymes, Intestinal Microflora, and Bacterial Translocation in Rats Subjected to Temporary Liver Inflow OcclusionTransplant. proc. 38378381
  202. 202. LyteM.1993The Role of Microbial Endocrinology in Infectious Disease.J. endocrinol. 137343345
  203. 203. FreestoneP. P.SandriniS. M.HaighR. D.LyteM.2008Microbial Endocrinology: How Stress Influences Susceptibility to InfectionTrends microbiol. 165564
  204. 204. FreestoneP. P.LyteM.CPNealMaggs. A. F.HaighR. D.WilliamsP. H.2000The Mammalian Neuroendocrine Hormone Norepinephrine Supplies Iron for Bacterial Growth in the Presence of Transferrin or LactoferrinJ. bacteriol. 18260916098
  205. 205. KinneyK. S.CEAustinMorton.DSSonnenfeldG.1999Catecholamine Enhancement of Aeromonas Hydrophila Growth.Microb. pathol. 268591
  206. 206. KinneyK. S.CEAustinMorton.DSSonnenfeldG.2000Norepinephrine as a Growth Stimulating Factor in Bacteria-Mechanistic Studies.Life sci. 6730753085
  207. 207. CPNealFreestone. P. P.MaggsA. F.HaighR. D.WilliamsP. H.LyteM.2001Catecholamine Inotropes as Growth Factors for Staphylococcus Epidermidis and Other Coagulase-Negative Staphylococci.FEMS microbiol. lett. 194163169
  208. 208. BelayT.SonnenfeldG.2002Differential Effects of Catecholamines on In Vitro Growth of Pathogenic Bacteria.Life sci. 71447456
  209. 209. BelayT.AvilesH.VanceM.FountainK.SonnenfeldG.2003Catecholamines and in vitro Growth of Pathogenic Bacteria: Enhancement of Growth Varies Greatly Among Bacterial Species.Life sci 7315271535
  210. 210. LyteM.ArulanandamB. P.FrankC. D.1996Production of Shigela-Like Toxins by Escherichia Coli O157:H7 can be Influenced by the Neuroendocrine Hormone Norepinephrine. J. lab. Clin. Med. 128392398
  211. 211. GreenB. T.LyteM.ChenC.XieY.MACasey-NarlaKulkarni.VulchanovaA.BrownL.D. R.2004Adrenergic Modulation of Escherichia Coli O157:H7 Adherence to the Colonic Mucosa.Am. j. physiol. gastrointest. liver physiol. 287: G1238G1246.
  212. 212. ChenC.BrownD. R.XieY.GreenB. T.LyteM.2003Catecholamines Modulate Escherichia Coli O157:H7 Adherence to Murine Cecal Mucosa.Shock. 20183188
  213. 213. ChenC.LyteM.StevensM. P.VulchanovaL.BrownD. R.2006Mucosally-Directed Adrenergic Nerves and Sympathomimetic Drugs Enhance Non-Intimate Adherence of Escherichia Coli O157:H7 to Porcine Cecum and ColonEur. j. pharmacol. 539116124
  214. 214. GreenB. T.LyteM.Kulkarni-NarlaA.BrownD. R.2003Neuromodulation of Enteropathogen Internalization in Peyer’s Patches from Porcine Jejunum.J. neuroimmunol. 1417482
  215. 215. Brown DR, Price LD2008Catecholamines and Sympathomimetic Drugs Decrease Early Salmonella Typhimurium Uptake into Porcine Peyer’s PatchesFEMS immunol. med. microbiol. 522935
  216. 216. KakunoY.HondaM.TakakuraK.1997Colonization Types of Escherichia Coli in Experimental Urinary Tract Infection in Compromised Mice Treated with Hydrocortisone. Kansenshogaku zasshi. 71652658
  217. 217. Schreiber KL, Brown DR2005Adrenocorticotrophic Hormone Modulates Escherichia Coli O157: H7 Adherence to Porcine Colonic Mucosa. Stress. 8185190
  218. 218. Sands BE2007Inflammatory Bowel Disease: Past, Present, and Future. J. gastroenterol. 4211625
  219. 219. FolignéB.NuttenS.SteidlerL.DenninV.GoudercourtD.MercenierA.PotB.2006Recommendations for Improved Use of The Murine TNBS-Induced Colitis Model in Evaluating Anti-İnflammatory Properties of Lactic Acid Bacteria: Technical and Microbiological Aspects. Dig. dis. sci. 51390400
  220. 220. CPTamboliNeut. C.DesreumauxP.ColombelJ. F.2004Dysbiosis in Inflammatory Bowel Disease Gut. 5314
  221. 221. ThomasL. V.OckhuizenT.2012New Insights into the Impact of the Intestinal Microbiota on Health and Disease: A Symposium ReportBr. j. nutr. 107: (Suppl 1): 1-13.
  222. 222. SteidlerL.2001Microbiological and Immunological Strategies for Treatment of Inflammatory Bowel Disease.Microbes infect. 311571166
  223. 223. ScanlanP. D.ShanahanF.O’MahonyC.MarchesiJ. R.2006Culture-Independent Analyses of Temporal Variation of The Dominant Fecal Microbiota and Targeted Bacterial Subgroups in Crohn’s DiseaseJ. clin. microbiol. 4439803988
  224. 224. FrankD. N.StAmand. A. L.FeldmanR. A.BoedekerE. C.HarpazN.PaceN. R.2007Molecular-Phylogenetic Characterization of Microbial Community Imbalances in Human Inflammatory Bowel Diseases.Proc. natl. acad. sci. 1041378013785
  225. 225. AlverdyJ.AoysE. (1991) The Effect of Glucocorticoid Administration on Bacterial Translocation. Ann. surg. 214 719723 .
  226. 226. SchiffrinE. J.CarterE. A.WalkerW. A.FriebergE.BenjaminJ.IsraelE. J.1993Influence of Prenatal Corticosteroids on Bacterial Colonization in the Newborn Rat.J. Pediatr. Gastroenterol. nutr. 17271275
  227. 227. DeitchE. A.WintertonJ.BergR.1987Effect of Starvation, Malnutrition and Trauma on the Gastrointestinal Tract Flora and Bacterial Translocation.Arch. Surg. 12210191024
  228. 228. Gorbach SL, Goldin BR1992Nutrition and the Gastrointestinal Microflora.Nutr. rev. 5037881
  229. 229. Saunders DR, Wiggins HS1981Conservation of Mannitol, Lactulose, and Raffinose by the Human Colon.Am. j. physiol. 241: G397G402.
  230. 230. Roberfroid MB2005Introducing Inulin-Type Fructans.Br. j. nutr. 93(Suppl 1): 13-25.
  231. 231. BSDrasarCrowther. J. S.GoddardP.HawksworthG.MJHillPeach. S.WilliamsR. E.RenwickA.1973The Relation between Diet and The Gut Microflora in Man. Proc. nutr. soc. 324952
  232. 232. GraceyM.SuharjonoSunoto.StoneD. E.1973Microbial Contamination of the Gut: Another Feature of Malnutrition.Am. j. clin. nutr. 2611701174
  233. 233. Finegold SM, Attebery HR, Sutter VL.1974Effect of Diet on Human Fecal Flora: Comparison of Japanese and American Diets.Am. j. clin. nutr. 271214561469
  234. 234. DeitchE. A.WintertonJ.LiM.BergR.1987The Gut as a Portal of Entry for Bacteremia. Role of Protein Malnutrition.Ann. surg. 205681692
  235. 235. MallettA. K.CABearneYoung. P. J.RowlandI. R.BerryC.1988Influence of Starches of Low Digestibility on the Rat Caecal Microflora.Br. j. nutr. 60597604
  236. 236. DeitchE. A.MaBergW. J. L.SpecianR. D.R. D.1990Protein Malnutrition Predisposes to Inflammatory-Induced Gut-Origin Septic States.Ann. surg. 211560568
  237. 237. HintonA.Jr BuhrR. J.IngramK. D.2000Physical, Chemical, and Microbiological Changes in the Crop of Broiler Chickens Subjected to Incremental Feed Withdrawal. Poult. sci. 79212218
  238. 238. Munro HN, Crim MC1988The Proteins and Amino Acids. In: Shils ME, Young VR editors. Modern nutrition in health and disease. Philadelphia: Lea & Febiger. 137
  239. 239. Viteri FE, Schenider RE1974Gastrointestinal Alterations in Protein-Calorie Malnutrition.Med. clin. n. am. 5814871505
  240. 240. HeyworthB.BrownJ.1975Jejunal Microflora in Malnourished Gambian Children.Arch. dis. child. 502733
  241. 241. Omoike IU, Abiodun PO1989Upper Small Intestinal Microflora in Diarrhea and Malnutrition in Nigerian Children.J. pediatr. gastroenterol. Nutr. 9314321
  242. 242. JirilloE.PasquettoN.MarcuccioL.MonnoR.De RinaldisP.FumarolaD.1975Endotoxemia Detected by Limulus Assay in Severe Malnourished Children. Plasma Effects on Leucocyte Migration: Preliminary Investigations.G. batteriol. virol. immunol. 68174178
  243. 243. Mc CowenK. C.LingP. R.CiccaroneA.MaoY.ChowJ. C.BistrianB. R.SmithR. J.2001Sustained Endotoxemia Leads to Marked Down-Regulation of Early Steps in the Insulin-Signaling Cascade.Crit. care med. 29839846
  244. 244. Schreiber RA, Walker WA1988The Gastrointestinal Barrier: Antigen Uptake and Perinatal Immunity.Ann. allergy. 61312
  245. 245. Sanderson IR, Walker WA1993Uptake and Transport of Macromolecules by the Intestine: Possible Role in Clinical Disorders (an Update).Gastroenterology104622639
  246. 246. StanghelliniV.BarbaraG.CremonC.CogliandroR.AntonucciA.GabusiV.FrisoniC.De GiorgioR.GrassoV.SerraM.CorinaldesiR.2010Gut Microbiota and Related Diseases: Clinical Features. Intern. emerg. med. 5 (Suppl 1): 57-63.
  247. 247. KatayamaM.XuD.SpecianR. D.DeitchE. A.1997Role of Bacterial Adherence and the Mucus Barrier on Bacterial Translocation: Effects of Protein Malnutrition and Endotoxin in Rats.Ann Surg. 225317326
  248. 248. AschkenasyA.1957On the Pathogenesis of Anemias and Leukopenias Induced by Dietary Protein Deficiency.Am. j. clin. nutr. 51425
  249. 249. TorunB.ViteriF. E.1989Nutrition and Function, with Emphasis on Physical Activity. In: Nutritional Problems of Children in the Developing World. M. Kretchmer M, Viteri FE, Falkner F, editors.
  250. 250. MarroquíL.BatistaT. M.GonzalezA.VieiraE.RafachoA.ColletaS. J.TabogaS. R.BoscheroA. C.NadalA.CarneiroE. M.QuesadaI.2012Functional and Structural Adaptations in the Pancreatic-Cell and Changes in Glucagon Signaling During Protein Malnutrition. Endocrinology. 15316631672

Written By

Hümeyra Ünsal and Muharrem Balkaya

Submitted: 23 April 2012 Published: 28 November 2012