ISSN: 2640-7612
Open Journal of Pediatrics and Child Health
Review Article       Open Access      Peer-Reviewed

Developmental significance of early gut-associated lymphoid tissue (GALT)-microbiota interactions in health and disease: Creating balance between tolerance and inflammation in children

Kaja Kasarello1 and Elzbieta M Sajdel-Sulkowska1,2*

1Departament of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw; 1b Banacha Street, 02-097 Warszawa, Poland
2Department of Psychiatry, Harvard Medical School, 75 Francis St, Boston, MA 02215, USA
*Corresponding author: Elzbieta M Sajdel-Sulkowska, Department of Psychiatry, Harvard Medical School, 75 Francis St, Boston, MA 02215, USA, E-mail: esulkowska@rics.bwh.harvard.edu
Received: 29 October, 2019 | Accepted: 15 November, 2019 | Published: 16 November, 2019
Keywords: Gastrointestinal Barrier (GIB); Gut symbiosis; Gut dysbiosis; Gut closure; Preterm infants; Inflammatory Bowel Disease (IBD); Necrotizing Colitis (NEC); Autism Spectrum Disorders (ASD); C-section

Cite this as

Kasarello K, Sajdel-Sulkowska EM (2019) Developmental significance of early gut-associated lymphoid tissue (GALT) - microbiota interactions in health and disease: creating balance between tolerance and inflammation in children. Open J Pediatr Child Health 4(1): 040-046. DOI: 10.17352/ojpch.000019

The establishment of gut microbiota in humans does not occur randomly but develops after birth through highly organized interactions between microorganisms, the immune processes orchestrated by the Gut-Associated Lymphoid Tissue (GALT), and a selective absorption to the blood regulated by the Gastrointestinal-Blood Barrier (GIB). In term infants, the initial colonization of gut microorganisms depends on the maturation of the GALT and critical closure of GIB and these interactions lead to the establishment of symbiotic conditions defined as a balance between immunity and infections. In preterm infants, development of the GALT is less complete at birth, and the GIB closure is delayed, both of which impact gut microbiota colonization resulting in dysbiosis. Early developmental dysbiosis may underlie non-infectious diseases such as allergies, autoimmune diseases, or inflammatory bowel diseases and contribute to the pathology of neurodevelopmental disorders. This review focuses on the developmental significance of GALT - microbiota interaction while comparing preterm and term infants. It concludes with the premiss that the developmental dysbiosis may have both short- and long-term impact on human health.

Introduction

The establishment of gut microbiota in humans commences at birth and involves highly organized interactions between microorganisms, the immune processes orchestrated by the Gut-Associated Lymphoid Tissue (GALT) and a selective absorption to the blood regulated by the Gastrointestinal Barrier (GIB). In term infants, the initial colonization of gut microorganisms depends on the maturation of the GALT and critical closure of GIB [1], and these interactions lead to the establishment of symbiotic conditions defined as a balance between immunity and infections. GALT is anatomically developed at birth in term infants but matures functionally through the interactions with microbiota [2]. The timing of the first gut closure occurs during the first 48 hours after birth decreasing GIB permeability [3]. In preterm infants, the anatomy of the GALT is less complete at birth, and the GIB closure is delayed [4], impacting gut microbiota colonization leading to dysbiosis.

This review summarizes the latest findings on GALTs functions, GALT-microbiota communication, and focuses on the developmental significance of GALT-microbiota interaction while comparing preterm and term infants. It concludes with a suggestion that the developmental dysbiosis may be a risk factor for such diseases as Necrotizing Colitis (NEC), Inflammatory Bowel Disease (IBD), autoimmune diseases, and neurodevelopmental disorders such as Autism Spectrum Disorders (ASD) and many others [5–7].

Gut-Associated Lymphoid Tissue (GALT): structure and functions

Intestinal lumen is part of the external environment that contains potentially harmful pathogens, such as bacteria, viruses or fungi. Gut epithelial cells act as a physical barrier between the intestinal lumen and the inner milieu preventing pathogens from entering the blood [8]. Tight-junction connections between epithelial cells provide mechanical protection against large molecules and microorganisms. Pathogens (e.g., bacteria, viruses) or their metabolites in the lumen may cause damage to the GI tract, eventually destroying the mechanical barrier allowing penetration of microorganisms into the bloodstream and underlying tissues. This points to the importance of proper recognition of antigens on commensal and pathogenic bacteria by the Pattern Recognition Receptors (PRRs) present on the surface of specialized cells of GALT and mounting the defense against pathogens.

GALT is composed of lymphoid tissue containing lymphocytes scattered in the epithelium and lamina propria, Peyer’s Patches (PPs), Isolated Lymphoid Follicles (ILFs), and M cells [9,10]. GALT is functionally connected to the mesenteric lymph nodes [8,11].

GALT functions include the identification of pathogens from non-pathogenic microorganisms or antigens and defense against pathogens [10]. The lumen of the human GI tract is colonized by a broad spectrum of symbiotic bacteria that plays a crucial role in the proper development and functioning of the organism. Furthermore, the intestines come in contact with dietary antigens, which are foreign to our immune system [8,12]. Evoking an immune response against such antigens would induce constant inflammation in the gut, causing chronic inflammatory diseases observed in food allergies or intolerances [13-15]. The phenomenon of suppression of immune response against non-harmful antigens from gut microbiota or food antigens is called oral tolerance [16,17] (Figure 1).

Antigen processing and induction of oral tolerance

Food- and microbiota-derived antigens present in the intestinal lumen, undergo endocytosis by M cells located in the gut epithelium and are transported to underlying tissue or to PPs, where they are processed by Antigen-Presenting Cells (APCs) such as Dendritic Cells (DCs) or macrophages. DCs also have the ability to extend their pseudopodia through the tight junctions to sample the antigens in the intestinal lumen [18,19]. APCs migrate to PPs or mesenteric lymph nodes and present processed antigen to naive T cells, which after acquiring the gut-homing ability, migrate to the lamina propria and epithelium [8,20,21]. Clones of the lymphocytes that reach the epithelium or lamina propria will be tolerant of the antigen previously presented by APCs.

Induction of tolerance is related to the dose of antigen presented. In response to low doses of antigen, naive T cells differentiate into regulatory T cells (Tregs) that produce anti-inflammatory cytokines, especially IL-4, IL-10, and TGF-β, which in turn, prevent inflammatory reaction (Th-1 dependent) induction. In response to high doses of antigens presented by APCs, T Cell Receptors (TCRs) are internalized or degraded, leading to the anergy or clonal deletion of lymphocytes [22,23]. Anergic lymphocytes are unresponsive to antigens and cannot migrate, creating the anti-inflammatory environment in the place of antigen presentation by APC [24]. Furthermore, the oral tolerance may be induced in the periphery by the population of migrating DCs, allowing the deletion of antigen-specific T cells (CD4+ and CD8+) in lymph nodes and liver [25-28].

What is the origin of this tolerance? The DCs in GALT, expressing CD103 have a tolerogenic phenotype, resulting from the exposure to TGF-β and retinoic acid produced by epithelial cells during their differentiation [29-31]. CD103+ DCs, in turn, produce anti-inflammatory cytokines, IL-10, and TGF-β that result in the differentiation of naive T cells into Th2 and Tregs and naive B lymphocytes into IgA producing B cells; these processes are also supported by TGF-β and retinoic acid produced by epithelial cells [32,33].

Recognition of the pathogen from non-pathogen

It has been suggested that the primary mechanism involved in distinguishing between symbiotic and pathogenic bacteria is based on the recognition of the Microorganism-Associated Molecular Patterns (MAMPs) by the pattern recognizing receptors (PRRs) on the surface of DCs. Binding the ligand to PRRs activates the intracellular signaling pathways, influencing the production of cytokines, and co-stimulatory molecules. Depending on the nature of the antigen, symbiotic or pathogenic, stimulation of PRRs results in the induction of tolerance or inflammation, respectively [34].

Role of the microbiota

Human intestines are colonized by the symbiotic bacteria belonging to about 500 - 1000 species, and in a number equivalent to the human cells; this number has been revised from the previous ratio of 10:1 [35]. Gut microbiota plays an essential role in maintaining the balance between tolerance to foreign antigens, and defense against foreign pathogens [36].

Toll-Like Receptors (TLR), belonging to PRRs, in general, are in general responsible for defense against pathogens, but in a tolerogenic environment in the gut, TLRs interact with symbiotic bacteria and do not evoke immune responses against them. Moreover, stimulation of the DCs’ TLRs results in the production of anti-inflammatory cytokine IL-10 [37,38], suggesting a direct interaction between immune cells and microbiota in inducing oral tolerance. Polysaccharide A (PSA) derived from Bacteroides fragilis induces differentiation of naive T cells into Treg subpopulation. PSA, by binding to TLR2 receptors on the surface of T cells, also induces the production of IL-10. Symbiotic bacteria use the TLR-dependent intracellular signaling pathway to inhibit the pro-inflammatory responses [39,40]. Indigenous Clostridium species increase the production of TGF-β and influences the accumulation of Tregs in intestines by increasing the level of secreted IL-10 [36]. Also, the metabolites of Clostridium species, short-chain fatty acids (SCFAs) promote the differentiation towards Tregs [41].

Microbiota may also influence other aspects of the gut immune response, such as migration of the DCs by regulating the release of chemokines. Immunomodulating mechanisms of microbiota also include elevation in IgA production, reduction of pro-inflammatory cytokines production, inhibition of lymphocyte proliferation and maintenance of gut epithelium integrity [23,42-44].

Responses to pathogenic microorganisms

Human GI plays a crucial role in the defense against pathogens that involves both specific and non-specific mechanisms. Mucus produced by goblet cells presents the physical barrier for pathogens. Enterocytes produce antimicrobial peptides (AMPs), which disrupt bacterial membranes leading to cell lysis or reduce the amount of the nutrients essential for bacterial cells through the process of so-called ‘nutritional immunity [45].

DCs of the GALT play an important role in the inflammatory response against pathogenic microorganisms. In the presence of inflammatory mediators, DCs produce the co-stimulatory molecules, which are essential for T-cell activation. During the inflammation, newly recruited DCs differentiate towards the pro-inflammatory phenotype, rather than the pro-tolerogenic. DCs that are recruited directly from the mesenteric lymph nodes are deprived of the influence of TGF-β and retinoic acid derived from the gut epithelium. Other immune cells residing in the GALT under the influence of the inflammatory mediators also change their phenotype into the proinflammatory one [46-48].

Regulation of GALT development: Establishing the immune balance in term infants

GALT is structurally developed in term infants, but after birth, undergoes the process of maturation in response to microorganisms colonizing the mucosal surfaces of the newborn [49]. Crosstalk between the immune system and microbiota is essential for the proper maturation of immune functions, establishing the Th1/Th2 balance, thus preventing Th1- or Th2-mediated diseases [2]. Once mature, to maintain gut homeostasis, GALT must maintain the established balance between Tregs and Th1/Th17 lymphocytes and assure the production of IgA by B cells, migrating from Peyer’s Patches to gut epithelium after contact with presented by APC antigen [41].

Immune development

The immune system consists of two distinct arms, innate and adaptive immunity. During inflammatory reactions, innate mechanisms are the first line of defense, followed by adaptive, antigen-specific responses. In newborns, the innate immune system presents the full repertoire of cells, such as neutrophils, monocytes, macrophages, DCs, natural killer cells, but compared to adults, their functions are depressed. Adaptive mechanisms are directed toward the maintenance of tolerance. T cells in newborns under the influence of foreign antigens differentiate into Th2 or Treg subtype, and B cells produce the low-affinity antibodies, both resulting in low-grade immune responses [50-52]. In the GALT of infants, immune reactions rely on the Innate Lymphoid Cells (ILCs), which have a similar function to T cells, but lack antigen-specificity. As adaptive mechanisms mature, CD4+ lymphocytes take over the function [53,54].

The anti-inflammatory characteristic of the immune reactions in neonates may be a life-saving adaptive mechanism in the fetus, allowing it to tolerate maternal-derived antigens. However, after birth, the suppressed and immature immune mechanisms contribute to the increased vulnerability of the newborns, relying on maternal antibodies received during fetal life [transplacental transfer] and breastfeeding [50-52].

Development of the epithelial barrier

Human fetal gut development begins early in the first trimester of gestation. During pregnancy, several processes resulting in the formation of structurally developed gut take place, including the growth, differentiation, and cell maturation, the formation of specific villi-crypt structures, and the elongation of intestines [55]. The Formation of tight-junctions begins with week 10 of gestation. Nevertheless, fetal gut remains permeable, allowing the exchange of molecules between fetal serum and amniotic fluid. Closure of the membrane, so-called “gut-closure” occurs during the first week of the postnatal period and is essential for the normal functioning of the intestinal barrier [3].

Breastmilk is vital for the proper gut-closure, and postnatal GALT maturation. Breastmilk contains many growth factors stimulating intestinal growth [56]. Nursing is also important for gut-closure, as significantly lower intestinal permeability was observed in breastfed newborns, compared to formula-fed children [57]. Furthermore, similar observations were made for preterm infants; those breastfed or receiving human milk had lower gut permeability than the formula-fed ones [58].

Studies performed using germ-free animals showed that commensal bacteria are also essential for gut maturation. Microbiota regulates the villus growth and proliferation of the epithelial cells [59].

Developmental disorders affecting the GIB barrier and resulting in increased intestinal permeability may contribute to the pathology of several diseases, such as NEC, infectious diarrhea, or allergic gastroenteropathy in newborns. It is also postulated that the disturbances in the gut barrier function during infancy may be a predisposing factor for inflammatory diseases such as inflammatory bowel diseases celiac disease or irritable bowel syndrome in adult life [60].

Regulation of the immune response by microbiota

The colonization of newborns GI tract by the mother’s vaginal bacteria during labor is essential for maturation of the gut immune system [61]. Research performed in germ-free mice showed strongly underdeveloped GALT, with a significantly reduced number of lymphocytes (both T cells and plasma cells) in lamina propria and hypoplastic PPs lacking most of the germinal centers. Interestingly, allowing the proper microbial colonization of germ-free animals reversed those changes, and caused proper maturation of GALT structures [62].

Microbiota exert diverse effects on the differentiation of T cells. Despite the tolerogenic environment in the intestines, the balance between Th1/Th17- and Th2-dependent response must be established and preserved [63,64]. Clostridium sp. induces the TGF-β production, and Tregs proliferation, helping to maintain the anti-inflammatory environment [65]. Segmented filamentous bacteria (SFB) induce Th17 cell differentiation, which is essential for defense against invading pathogens [66,67]. Also, the presence of Th1/Th17 cells and inflammatory mediators (IL-17, IL-22) in the gut is vital, while inhibiting excessive Th2-dependent mechanisms expansion is relevant in preventing asthma, allergic reactions, and other Th2-dependent diseases development. In children delivered by C-section, there is a low diversity and abundance in bacteria species colonizing the GI tract, a very low number of Bacteroides and Bifidobacterium species, and reduced levels of mediators of the Th1-dependent responses. This, in turn, shifts the balance towards the Th2-mediated mechanisms [68,69]. On the other hand, preventing excessive Th17-dependent responses mediating the autoimmunity is necessary to suppress autoimmune diseases, such as rheumatoid arthritis or multiple sclerosis [43,70].

Microbiota influences editing the B cell receptors and shaping its repertoire [71]. Additionally, symbiotic bacteria promote the differentiation of Bregs producing IL-10, helping to maintain the gut-homeostasis [72]. In germ-free mice, the increased levels of IgE, are accompanied by the decreased IgG and IgA levels [73]. Microbiota transplant to the germ-free mice results in increased production of both IgG and IgA [74].

Microbiota regulation of immune development involves the epigenetic mechanisms, such as DNA methylation or histone phosphorylation and acetylation, involved in gene expression and silencing [75,76]. It has been proposed that the insufficient early-life gut colonization by microbiota may result in silencing the IFN-γ genes in naive T cells, shifting the balance in Th1/Th17- and Th2-dependent mechanisms towards Th2, which may result in an allergic response [77]. Immune responses may also be influenced by epigenetic modifications of nuclear factor-κB, TLRs, and many other genes of molecules involved in the modulation of inflammatory processes [78].

Interestingly, a recent report showing the presence of symbiotic bacteria in the placenta, amniotic fluid, and meconium [79,80] challenged the hypothesis of fetal gut sterility. Nevertheless, the data showed a very low diversity and abundance of bacteria [61,81]. However, a critical assessment of the evidence supporting these two opposing hypotheses, pointing to the methodological errors and the ability to reliably derive axenic animals via cesarean sections, strongly supports sterility of the fetal environment in mammals [82].

GALT development in preterm infants

As discussed above, the crosstalk between gut microbiota and GALT is essential for the proper development and functioning of the organism. However, there are several factors that may interfere with these processes [6]. Depending on the gestational age at the time of birth, the immune system may be more or less mature, and the degree of prematurity causes abnormal response for commensal bacteria evoking the inflammatory response in place of immune tolerance. The timing of GIB closure is delayed in preterm infants and the prematurity of the GIB barrier contributes to the overall pathological state. The underdeveloped physical epithelial barrier in terms of abnormal cell proliferation, disturbed cell differentiation, or tight-junction formation leads to antigen penetration and inflammation [83]. Inflammatory reactions in intestines lead to the damage of the epithelium and disruption of the gut-blood barrier, promoting bacteria penetration into underlying tissues, eventually causing life-threatening conditions like sepsis or NEC [4,84].

Prematurity implies a further risk of gut-microbiota dysbiosis. Preterm children are often delivered by C-section that results in gut colonization by the skin bacteria, instead of vaginal and fecal microorganisms during normal labor. Secondly, preterm infants are often formula-fed, deprived of mother’s milk containing nutrients used by microbiota, and also bacteria that colonize the intestines of the infant. Additionally, treatment with antibiotics and the sanitary conditions of intensive care nurseries reduce the diversity of microorganisms colonizing the gut and further contribute to the microbiota dysbiosis in the newborn [69,85]

Summary

This review provides evidence supporting the concept of underdeveloped functions of GALT, dysfunctional GIB, and altered GALT microbiota interactions in preterm infants. GALT is not only critical during development, determining the tolerance and supporting immune function. It also plays an essential role as a gate-keeper in maintaining a healthy GIB barrier between the external environment and the human organism. Developmental GALT dysregulation, GIB barrier dysfunction, and dysbiosis may all contribute to non-infectious diseases, such as autoimmune diseases, allergies, or other inflammatory diseases. Further studies of the mechanisms involved in the establishment of developmental gut symbiosis are crucial for the proper care of preterm infants and lowering long-term health risks.

This project was supported by the CePT infrastructure financed by the European Regional Development Fund with the Operational Programme “Innovative Economy” for 2007-2013.

  1. Makowska M, Kasarello K, Bialy M, Sajdel-Sulkowska EM (2016) Autism: “Leaky Gut”, prematurity and lactoferrin. Austin J Autism Relat Disabil 2: 1021. Link: http://bit.ly/2CIF2zl
  2. Forchielli ML, Walker WA (2005) The role of gut-associated lymphoid tissues and mucosal defence. Br J Nutr 93: S41-S48. Link: http://bit.ly/2q5O3jB
  3. Maheshwari A, Zemlin M (2009) Ontogeny of the intestinal immune system. Nephrol Rev 2: 18-26. Link:  http://bit.ly/374qKXU
  4. Groer MW, Luciano AA, Dishaw LJ, Ashmeade TL, Miller E, et al. (2014) Development of the preterm infant gut microbiome: a research priority. Microbiome 2: 38. Link: http://bit.ly/2NOVEMw  
  5. Koboziev I, Karlsson F, Grisham MB (2010) Gut-associated lymphoid tissue, T cell trafficking, and chronic intestinal inflammation. Ann NY Acad Sci 1207: E86- E93. Link: http://bit.ly/2NPAiP5
  6. Neu J (2007) Perinatal and neonatal manipulation of the intestinal microbiome: A note of caution. Nutr Rev 65: 282-285. Link: http://bit.ly/2NNRGDw  
  7. Yoo BB, Mazmanian SK (2017) The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity 46: 910-926. Link: http://bit.ly/33KrjUJ
  8. Reis BS, Mucida D (2012) The role of the intestinal context in the generation of tolerance and inflammation. Clin Dev Immunol 2012: 157948. Link: http://bit.ly/2rJPowJ   
  9. Neutra MR, Frey A, Kraehenbuhl JP (1996) Epithelial M cells: Gateways for mucosal infection and immunization. Cell 86: 345-348. Link: http://bit.ly/2NNCzKh  
  10. Nagler-Anderson C (2001) Man the barrier! Strategic defences in the intestinal mucosa. Na tRev Immunol 1: 59-67. Link: http://bit.ly/32PKLOB  
  11. Bain CC, Mowat AM (2014) Macrophages in intestinal homeostasis and inflammation. Immunol Rev 260: 102-117. Link:  http://bit.ly/2NNCYMN
  12. Ménard S, Cerf-Bensussan N, Heyman M (2010) Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal Immunol 3: 247-259. Link: http://bit.ly/37h4xGi  
  13. Faria AMC, Weiner HL (2006) Oral tolerance: therapeutic implications for autoimmune diseases. Clin Dev Immunol 13: 143-157. Link: http://bit.ly/33PGKLq  
  14. Weiner HL, da Cunha AP, Quintana F, Wu HY (2011) Oral tolerance. Immunol Res 241: 241-259. Link: http://bit.ly/32Niw37  
  15. Pabst O, Mowat AM (2012) Oral tolerance to food protein. Mucosal Immunol 5: 232-239. Link: https://go.nature.com/379l4fm
  16. Wells HG (1911) Studies on the chemistry of anaphylaxis (iii). experiments with isolated proteins, especially those of the hen’s egg. J Infect Dis 9: 147-171.  Link: http://bit.ly/2CJAWaa
  17. Weiner HL (1997) Oral tolerance: Immune mechanisms and treatment of autoimmune diseases. Immunol Today 18: 335-343. Link: http://bit.ly/351cI7A  
  18. Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, et al. (2001) Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol 2: 361-367.  Link: http://bit.ly/32Om2dx
  19. Farache J, Koren I, Milo I, Gurevich I, Kim KW, et al. (2013) Luminal Bacteria Recruit CD103+ Dendritic Cells into the Intestinal Epithelium to Sample Bacterial Antigens for Presentation. Immunity 38: 581-595.  Link: http://bit.ly/32NrOfm
  20. Tordesillas L, Berin MC (2018) Mechanisms of Oral Tolerance. Clin Rev Allergy Immunol 55: 107-177.  Link: http://bit.ly/37a3wzu
  21. Geem D, Ngo V, Harusato A, Chassaing B, Gewirtz AT, et al.  (2016) Contribution of Mesenteric Lymph Nodes and GALT to the Intestinal Foxp3+ Regulatory T-Cell Compartment. Cell Mol Gastroenterol Hepatol 2: 274-280.  Link: http://bit.ly/374udFU
  22. Benson JM, Campbell KA, Guan Z, Gienapp IE, Stuckman SS, et al. (2000) T-cell activation and receptor downmodulation precede deletion induced by mucosally administered antigen. J Clin Invest 106: 1031-1038. Link: http://bit.ly/2COwy9S  
  23. Wells JM, Rossi O, Meijerink M, van Baarlen P (2011) Epithelial crosstalk at the microbiota-mucosal interface. Proc Natl Acad Sci 108: 4607-4614. Link: http://bit.ly/2OdQSqz
  24. Mirenda V, Millington O, Lechler RI, Scott D, Hernandez-Fuentes MP, et al. (2005) Tolerant T cells display impaired trafficking ability. Eur J Immunol 35: 2146-2156.  Link: http://bit.ly/2rOqj41
  25. Song F, Guan Z, Gienapp IE, Shawler T, Benson J, et al. (2006) The thymus plays a role in oral tolerance in experimental autoimmune encephalomyelitis. J Immunol 177: 1500-1509. Link: http://bit.ly/32PNcRf  
  26. Song F, Wardrop RM, Gienapp IE, Stuckman SS, Meyer AL, et al. (2008) The Peyer’s patch is a critical immunoregulatory site for mucosal tolerance in experimental autoimmune encephalomylelitis (EAE). J Autoimmun 30: 230-237. Link: http://bit.ly/2Xf99YK
  27. Goubier A, Dubois B, Gheit H, Joubert G, Villard-Truc F, et al. (2008) Plasmacytoid Dendritic Cells Mediate Oral Tolerance. Immunity 29: 464-475. Link: http://bit.ly/2CHrsMV  
  28. Idoyaga J, Fiorese C, Zbytnuik L, Lubkin A, Miller J, et al. (2013) Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Invest 123: 844-854. Link: http://bit.ly/2QkGQ9Z
  29. Annacker O, Coombes JL, Malmstrom V, Uhlig HH, Bourne T, et al. (2005) Essential role for CD103 in the T cell–mediated regulation of experimental colitis. J Exp Med 202: 1051-1061. Link: http://bit.ly/32PeZRC
  30. Coombes JL, Siddiqui KRR, Arancibia-Cárcamo CV, Hall J, Sun CM, et al. (2007) A functionally specialized population of mucosal CD103 + DCs induces Foxp3 + regulatory T cells via a TGF-β– and retinoic acid–dependent mechanism. J Exp Med 204: 1757-1764. Link: http://bit.ly/2CNa8Gh
  31. Iliev ID, Mileti E, Matteoli G, Chieppa M, Rescigno M (2009) Intestinal epithelial cells promote colitis-protective regulatory T-cell differentiation through dendritic cell conditioning. Mucosal Immunol 2: 340-350. Link: http://bit.ly/2NPmIeo  
  32. Iwasaki A, Kelsall BL (1999) Freshly isolated Peyer’s patch, but not spleen, dendritic cells produce interleukin 10 and induce the differentiation of T helper type 2 cells. J Exp Med 190: 229-239.  Link: http://bit.ly/2pjANqX
  33. Uto T, Takagi H, Fukaya T, Nasu J, Fukui T, et al. (2018) Critical role of plasmacytoid dendritic cells in induction of oral tolerance. J Allergy Clin Immunol 141: 2156-2167. Link: http://bit.ly/2KnS9ud
  34. Bergstrom KS, Sham HP, Zarepour M, Vallance BA (2012) Innate host responses to enteric bacterial pathogens: a balancing act between resistance and tolerance. Cell Microbiol 14: 475-484. Link: http://bit.ly/32L1mD7
  35. Sender R, Fuchs S, Milo R (2016) Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol 14: e1002533. Link: http://bit.ly/2XnI2uM
  36. Atarashi K, Honda K (2011) Microbiota in autoimmunity and tolerance. Curr Opin Immunol 23: 761-768. Link: http://bit.ly/2Xge7o4
  37. McLoughlin RM, Mills KHG (2011) Influence of gastrointestinal commensal bacteria on the immune responses that mediate allergy and asthma. J Allergy Clin Immunol 127: 1097-1107. Link: http://bit.ly/2QlecFK
  38. Sansonetti PJ (2011) To be or not to be a pathogen: That is the mucosally relevant question. Mucosal Immunol 4: 8-14. Link: http://bit.ly/2q5R03H  
  39. Ochoa-Repáraz J, Mielcarz DW, Wang Y, Begum-Haque S, Dasgupta S, et al. (2010) A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol 3: 487-495. Link: http://bit.ly/2qSIMfe
  40. Round JL, Lee SM, Li J, Tran G, Jabri B, et al. (2011) The Toll-like receptor pathway establishes commensal gut colonization. Science 332: 974–977. Link: http://bit.ly/2Krp3dw  
  41. Yang H, Duan Z (2018) The Local Defender and Functional Mediator: Gut Microbiome. Digestion 97: 137-145. Link: http://bit.ly/2Ko5eU6
  42. Isolauri E, Sütas Y, Kankaanpää P, Arvilommi H, Salminen S, et al. (2001) Probiotics: Effects on immunity. Am J Clin Nutr 73: 444S-450S. Link: http://bit.ly/2CQRddr  
  43. Lee YK, Mazmanian SK (2010) Has the microbiota played a critical role in the evolution of the adaptive immune system? Science 330: 1768-1773. Link: http://bit.ly/2XhVct8
  44. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, et al. (2014) Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci 111: 13145-13150. Link: http://bit.ly/33P6Az9
  45. Chung LK, Raffatellu M (2018) G.I. Pros: Antimicrobial defense in the gastrointestinal tract. Semin Cell Dev Biol 88: 129-137.  Link: http://bit.ly/2Qiy6ku
  46. Strobel S, Mowat AM (1998) Immune responses to dietary antigens: Oral tolerance. Immunol Today. 19: 173-181. Link: http://bit.ly/2Qh8JQe  
  47. Mowat AM (2005) Dendritic cells and immune responses to orally administered antigens. Vaccine 23: 1797-1799. Link: http://bit.ly/32OXd1d  
  48. Rescigno M (2011) Dendritic cells in oral tolerance in the gut. Cell Microbiol 13: 1312-1318.  Link: http://bit.ly/2XdIg7C
  49. Olin A, Henckel E, Chen Y, Lakshmikanth T, Pou C, et al. (2018) Stereotypic Immune System Development in Newborn Children. Cell 174: 1277-1292. Link: http://bit.ly/2CR12YR
  50. Simon AK, Hollander GA, McMichael A (2015) Evolution of the immune system in humans from infancy to old age. Proc R Soc B Biol Sci 282: 20143085. Link: http://bit.ly/32StDHP  
  51. Fouda GG, Martinez DR, Swamy GK, Permar SR (2018) The Impact of IgG Transplacental Transfer on Early Life Immunity. Immunohorizons 2: 14-25. Link: http://bit.ly/377BrsC  
  52. Prescott SL, Macaubas C, Holt BJ, Smallacombe TB, Loh R, et al. (1998) Transplacental priming of the human immune system to environmental allergens: universal skewing of initial T cell responses toward the Th2 cytokine profile. J Immunol 160: 4730–4737. Link: http://bit.ly/32LIFz5
  53. Mao K, Baptista AP, Tamoutounour S, Zhuang L, Bouladoux N, et al. (2018) Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid metabolism. Nature 554: 255-259. Link: http://bit.ly/2CJF4al
  54. Das A, Harly C, Yang Q, Bhandoola A (2018) Lineage specification in innate lymphocytes. Cytokine Growth Factor Rev 42: 20-26.  Link: http://bit.ly/2Oil97X
  55. Montgomery RK, Mulberg AE, Grand RJ (1999) Development of the human gastrointestinal tract: Twenty years of progress. Gastroenterology 116: 702-731. Link: http://bit.ly/32U0SuJ  
  56. Cummins AG, Thompson FM (2002) Effect of breast milk and weaning on epithelial growth of the small intestine in humans. Gut 51: 748-754. Link: http://bit.ly/33PKbBP
  57. Catassi C, Bonucci A, Coppa GV, Carlucci A, Giorgi PL (1995) Intestinal permeability changes during the first month: effect of natural versus artificial feeding. J Pediatr Gastroenterol Nutr 21: 383-386. Link: http://bit.ly/32RDSwa  
  58. Taylor SN, Basile LA, Ebeling M, Wagner CL (2009) Intestinal Permeability in Preterm Infants by Feeding Type: Mother’s Milk Versus Formula. Breastfeed Med 4: 11-15. Link: http://bit.ly/2OjxE2O  
  59. Louis NA, Lin PW (2009) The Intestinal Immune Barrier. Neo Reviews 10: e180– e190. Link: http://bit.ly/2XmhzO3
  60. Anderson RC, Dalziel JE, Gopal P, Bassett S, Ellis A, et al. (2009) The Role of Intestinal Barrier Function in Early Life in the Development of Colitis. London (UK). Link: http://bit.ly/33SQuEw
  61. Zhao Q, Elson CO (2018) Adaptive immune education by gut microbiota antigens. Immunology 154: 28-37. Link: http://bit.ly/2QlX3f9  
  62. Macpherson AJ, Harris NL (2004) Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol 4: 478-485. Link: http://bit.ly/2KppPHK
  63. Yu Q, Jia A, Li Y, Bi Y, Liu G (2018) Microbiota regulate the development and function of the immune cells. Int Rev Immunol 37: 79-89. Link: http://bit.ly/379e9mc
  64. Gaboriau-Routhiau V, Rakotobe S, Lécuyer E, Mulder I, Lan A, et al. (2009) The Key Role of Segmented Filamentous Bacteria in the Coordinated Maturation of Gut Helper T Cell Responses. Immunity 31: 677–689. Link: http://bit.ly/33Nvnn6
  65. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, et al. (2011) Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species. Science 331: 337-341. Link: http://bit.ly/2QmLrIS  
  66. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, et al. (2009) Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria. Cell 139: 485-498.  Link: http://bit.ly/2Kps8uv
  67. Ivanov II, Frutos R de L, Manel N, Yoshinaga K, Rifkin DB, et al. (2008) Specific Microbiota Direct the Differentiation of IL-17-Producing T-Helper Cells in the Mucosa of the Small Intestine. Cell Host Microbe 4: 337–349. Link: http://bit.ly/355cLiQ  
  68. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, et al. (2014) Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by Caesarean section. Gut 63: 559-566. Link: http://bit.ly/2poS6Hk  
  69. Ximenez C, Torres J (2017) Development of the Microbiota in Infants and its Role in Maturation of the Gut Mucosa and the Immune System. Arch Med Res 48: 666-680. Link: http://bit.ly/2NQ4MAr
  70. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, et al. (2010) Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32: 815-827. Link: http://bit.ly/2CHqOyX  
  71. Wesemann DR, Portuguese AJ, Meyers RM, Gallagher MP, Cluff-Jones K, et al. (2013) Microbial colonization influences early B-lineage development in the gut lamina propria. Nature 501: 112–115. Link: http://bit.ly/375XFvb  
  72. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, et al. (2014) Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat Med 20: 1334–1339. Link: http://bit.ly/379fAB6
  73. Gensollen T, Blumberg RS (2017) Correlation between early-life regulation of the immune system by microbiota and allergy development. J Allergy Clin Immunol 139: 1084-1091. Link: http://bit.ly/2CN05km
  74. Kim CH (2016) B cell-helping functions of gut microbial metabolites. Microb Cell 3: 529–531. Link: http://bit.ly/2CLHKnP
  75. Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, et al. (2016) Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol Cell 64: 982–992. Link: http://bit.ly/2CMEbOk  
  76. Paul B, Barnes S, Demark-Wahnefried W, Morrow C, Salvador C, et al. (2015) Influences of diet and the gut microbiome on epigenetic modulation in cancer and other diseases. Clin Epigenetics 7: 112.  Link: http://bit.ly/3552fIq
  77. Vuillermin PJ, Ponsonby AL, Saffery R, Tang ML, Ellis JA, et al.  (2009) Microbial exposure, interferon gamma gene demethylation in naive T-cells, and the risk of allergic disease. Allergy 64: 348-353.  Link: http://bit.ly/2pmhIEL
  78. Indrio F, Martini S, Francavilla R, Corvaglia L, Cristofori F, et al. (2017) Epigenetic Matters: The Link between Early Nutrition, Microbiome, and Long-term Health Development. Front Pediatr 5: 178. Link: http://bit.ly/33PYp5G
  79. Walker RW, Clemente JC, Peter I, Loos RJF, et al. (2017) The prenatal gut microbiome: are we colonized with bacteria in utero? Pediatr Obes 12: 3–17. Link: http://bit.ly/2QkRMUY
  80. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, et al. (2014) The placenta harbors a unique microbiome. Sci Transl Med 6: 237. Link: http://bit.ly/379hpxW
  81. Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, et al. (2015) The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Heal Dis 26: 26050. Link: http://bit.ly/2QiNZYa
  82. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J (2017) A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: Implications for research on the pioneer infant microbiome. Microbiome 5: 48. Link: http://bit.ly/2Kpqw3N  
  83. Kelly JR, Kennedy PJ, Cryan JF, Dinan TG1, Clarke G, et al. (2015) Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci  9: 392. Link: http://bit.ly/2rHY4nf
  84. Hunter CJ, De Plaen IG (2014) Inflammatory signaling in NEC: Role of NF-κB, cytokines and other inflammatory mediators. Pathophysiology 21: 55–65. Link: http://bit.ly/32Ihq8M  
  85. Tasnim N, Abulizi N, Pither J, Hart MM, Gibson DL, et al. (2017) Linking the gut microbial ecosystem with the environment: Does gut health depend on where we live? Front Microbiol 8: 1935.  Link: http://bit.ly/2OdoftM
© 2019 Kasarello K, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
 

Help ?