1,440
Views
7
CrossRef citations to date
0
Altmetric
Reviews

Modulation of the immune system by the gut microbiota in the development of type 1 diabetes

ORCID Icon, , ORCID Icon & ORCID Icon
Pages 2580-2596 | Received 12 Jun 2018, Accepted 17 Aug 2018, Published online: 19 Sep 2018

ABSTRACT

T1D is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing β-cells in the pancreatic islets of Langerhans, resulting in hyperglycemia, with patients requiring lifelong insulin treatment. Many studies have shown that genetics alone are not sufficient for the increase in T1D incidence and thus other factors have been suggested to modify the disease risk. T1D incidence has sharply increased in the developed world, especially amongst youth. In Europe, T1D incidence is increasing at an annual rate of 3–4%. Increasing evidence shows that gut microbiota, as one of the environmental factors influencing diabetes development, play an important role in development of T1D. Here, we summarize the current knowledge about the relationship between the microbiota and T1D. We also discuss the possibility of T1D prevention by changing the composition of gut microbiota.

This article is part of the following collections:
Immunotherapy of Autoimmune Diseases

Introduction

The elucidation of the complex interactions between the gut microbiota, metabolism, and the immune system may lead to groundbreaking changes as to how specific diseases are prevented and treated. The gut microbiota refers to the community of bacteria located within the intestine that have coevolved through millions of years with the host. This symbiotic relationship is important for many host functions including digestion, nutrient acquisition, and the development of the immune system.Citation1

The gut microbiota encodes trillions of genes, of which approximately 5–10 million are unique.Citation2-Citation4 In total, there are ~ 150 times more genes than in the human genome.Citation3 In healthy humans, the number of the bacteria increases exponentially from the small intestine to the colon; thus, the colon is the main contributor to the total bacterial population in the gut.Citation5 The main commensal bacterial phlya in the gut microbiota include, Actinobacteria, Bacteroidetes, Firmicutes, Proteobacteria, and Verrucomicrobia with the vast majority consisting of Bacteroidetes and Firmicutes.

The gut microbiota of each individual host is diverse and unique.Citation6 For maintenance of good health, there is a natural balance between the host and its microbial community. However, dysbiosis, which is a disturbance in the balance between the host and the microbial community, is associated with various chronic diseases, including obesity, inflammatory bowel disease (IBD), type 1 diabetes (T1D) and type 2 diabetes (T2D).Citation7

Recent evidence suggests the gut microbiota may begin colonization in utero as bacteria have been detected in the intrauterine environment including the amniotic fluid,Citation8 placenta,Citation9 meconium,Citation10 and the umbilical cord.Citation11 While the majority of these studies utilize 16S rRNA sequencing or PCR, in a small number the investigators have grown bacteria by culturing the amniotic fluidCitation8 or the umbilical cordCitation11 samples in different bacterial culture conditions. In addition, the development of the gut microbiota can be influenced by the delivery mode. Infants delivered by Caesarean section (C-section) exhibit less bacterial diversity up to 2 years after birth compared with those delivered vaginally.Citation12 C-section deliveries have also been associated with an increased risk of obesityCitation13 or T1DCitation14 later in life, which may be linked to the fact that the gut microbiota continue to develop into adulthood.Citation15 This potentially provides a greater window of opportunity for therapeutic modulation of the gut microbiota.

There are many ways that the gut microbiota can be modified, including the environments we live in (e.g. rural or urban), diet and food supplements (probiotics), the use of antibiotics or other medications during illness. Other factors, such as age and gender, can also modify the microbial composition over time. For example, the healthy pediatric gut microbiota exhibit significant compositional and functional differences from those of adults.Citation15 It has been shown that children had increased abundances of Bifidobacterium, Faecalibacterium, and Lachnospiraceae compared with adults, while adults had increased abundance of Bacteroides.Citation1 Currently, it is not clear what constitutes a “healthy” gut microbial composition; however, the microbiota are indispensible for the digestion of nutrients,Citation16 development of mucosal immunity,Citation17 and supporting gut-brain communication.Citation18 Furthermore, a loss of diversity in the gut microbiota or alterations in microbial functions have been associated with risk of developing chronic diseases including T1D and other autoimmune or inflammatory disorders. Therefore, developing or retaining a “healthy” microbiota is important.

T1D is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing β-cells in the pancreatic islets of Langerhans, resulting in hyperglycemia, with patients requiring lifelong insulin treatment. Many studies have shown that genetics alone are not sufficient for the increase in T1D incidence and thus other factors have been suggested to modify the disease risk. T1D incidence has sharply increased in the developed world, especially amongst youth.Citation19 In Europe, T1D incidence is increasing at an annual rate of 3–4%.Citation20 Increasing evidence shows that gut microbiota, as one of the environmental factors influencing diabetes development, play an important role in development of T1D.Citation21 Here, we summarize the current knowledge about the relationship between the microbiota and T1D. We also discuss the possibility of T1D prevention by changing the composition of gut microbiota.

Animal models of T1D

To gain the best knowledge of the mechanism(s) of disease development, the ideal studies are in vivo investigations. For ethical reasons, there are considerable limitations to in vivo studies in humans. However, animal models of human diseases provide an alternative system to investigate the mechanism behind the immune response within the pancreas, in the case of T1D, or the bacteria in the gut, in vivo, to answer questions that we cannot do in humans. Utilizing bench to bedside and bedside to bench approaches can further expand our understanding and help us to achieve the ultimate goal of preventing T1D development or to develop a cure.

There are two widely used rodent models for human T1D research – the non-obese diabetic (NOD) mouseCitation22 and the bio-breeding (BB) rat.Citation23 Both the NOD mouse and the BB rat develop spontaneous T1D, similar to humans. NOD mice usually develop T1D after 10 weeks of age,Citation24 while BB rats develop T1D from 7 to 14 weeks.Citation25 In affected humans, the age of onset of T1D typically ranges from 6 months to late adolescence. The NOD mouse and the BB rat also carry the T1D susceptibility major histocompatibility complex (MHC) class II genes, similar to the human T1D susceptibility MHCII alleles. The NOD mouse recapitulates many features of human T1D, especially those T1D susceptibility genes.Citation26 However, the pathogenesis of T1D in both humans and animal models is not solely determined by genetics; the disease onset is influenced by a combination of genetic and environmental factors. Pathologically, humans,Citation27 the BB ratCitation28 and NOD miceCitation29 display infiltration of lymphocytes in the pancreas, namely, insulitis. The immune cells that are involved in the destruction of insulin-producing beta cells in humans, are similar to those cells present in NOD mice and BB rats during diabetes development. These cells are mainly autoreactive T cells as T1D is a T cell-mediated disease. Furthermore, the autoantigens that the T cell recognizes in human T1D are also present in the NOD mouse.Citation30 In humans, a gender bias emerges after puberty with a small increase in the number of affected males,Citation31 while the majority of diabetic NOD mice are females.Citation22 However, there is no gender bias in T1D development in BB rats.Citation23

Unlike human studies, studies using animal models can be better controlled in order to minimize variables and assess the effect of different environmental factors, such as diet, mode of birth delivery and usage of medication, on the gut microbiota and the development of T1D. Therefore, studies using animal models provide an extremely valuable and unique tool for gaining more insightful knowledge about the disease. In addition, humansCitation32 and rodentsCitation33,Citation34 with T1D have been shown to exhibit similar gastroenterological abnormalities, including increased intestinal permeability, altered microvilli, leaky tight junctions, and altered gut microbiota.Citation35-Citation37 Similar to humans, Bacteroidetes and Firmicutes are also the dominant phyla in relation to the composition of gut microbiota. However, there are also major differences as 85% of the bacterial genera found in mice are absent in humans.Citation38 Moreover, a disadvantage of well-controlled studies using in-bred animal models may be a lack of direct translation to humans, who are extremely heterogeneous.

Gut microbiota and T1D

Hygiene hypothesis

The concept of the gut microbiota as a major environmental factor influencing T1D supports the rising incidence rates in developed countries. The hygiene hypothesis was originally proposed in relation to observations of respiratory problems, hygiene and household size.Citation39 A modification of this may help to explain the increased T1D incidence as a result of reduced diversity in the microbiota. The sharp increase in T1D incidence dates back to the mid 20th century where children were raised in environments with increased levels of sanitation and thus have less exposure to bacteria and parasites. The hygiene hypothesis has been tested in NOD mice, as the cleaner the living conditions, the higher the incidence of diabetes found in NOD mice.Citation40 Moreover, studies have found that infection of NOD mice early in life with a number of different bacteria can prevent T1D.Citation41,Citation42 Human epidemiological studies showed that the incidence of T1D and allergies is much lower in developing countries where the living standard is low but the rate of bacterial or parasite infection is high.Citation43 Links found between gut microbiota and T1D, discussed in this review, have prompted questions on how the gut microbiota can be modulated in order to alter T1D development.

Gut microbiota in T1D

Both the gut microbiota composition and the immune system co-evolve and develop together over time, with young children exhibiting reduced microbial diversity and a less mature immune system compared to adults.Citation15,Citation44 Therefore, it is important to understand how the gut microbiota interacts with the immune system and further, how these interactions alter susceptibility to T1D. Roesch and colleagues found that bacteria of the Bacteroides genus were more common in diabetes-prone BB rats (BB-DP) than they were in diabetes-resistant BB rats (BB-DR).Citation45 However, the abundance of bacteria belonging to the Lactobacillus and Bifidobacterium genera was higher in BB-DR rats than in BB-DP rats.Citation45 Altered microbiota were also found between the NOD mouse and non-obese diabetes resistant (NOR) mouse.Citation46 According to Daft and colleagues, the NOD mouse has a lower Firmicutes:Bacteroidetes ratio as well as a lower abundance of Prevotella compared to the NOR mouse.Citation46 This profile is also seen in children with T1D compared to age-matched healthy children.Citation37 Long-term changes in the gut microbiota of NOD or NOR mice can be accomplished by cross-fostering, whereby NOD mice are nursed by NOR mothers and vice versa.Citation46 Cross-fostering of NOD mice results in both the loss of some diabetogenic bacteria and the gain of bacteria associated with diabetes protection. Further, NOD mice fostered by NOR mothers had a decreased incidence of T1D.Citation46

Interactions of gut microbiota with known T1D susceptibility loci can influence the risk of developing T1D. Genetic susceptibility at the MHC loci is the most important risk factor for T1D development in both NOD mice and humans. NOD mice express the gene encoding MHC class II IAg7, which is a homolog of human HLA-DQ8.Citation47-Citation49 NOD mice do not express IE, another MHC class II gene, a homolog of human HLA-DR.Citation48,Citation49 Expression of IE, or expression of IAb, the MHC class II locus for C57BL/6 mice, instead of IAg7, is associated with protection from disease in NOD mice.Citation50,Citation51 Interestingly, a recent study by Silverman et al. suggested the mechanism of disease protection is mediated by gut microbiota in IE-expressing NOD mice.Citation52 The authors showed that expression of the IE transgene results in the compositional changes in gut microbiota which contributes to T1D protection. To prove that the disease protection was indeed mediated by the gut microbiota, the authors treated the IE transgenic NOD mice with different antibiotics. Administration of vancomycin or metronidazole, but not neomycin or ampicillin, in the drinking water, disturbed the gut microbiota sufficiently to induce insulitis in the normally insulitis-free IE transgenic NOD mice.Citation52 Although the presence of IAb in NOD mice was also able to alter the gut microbiota composition, the effect on diabetes development was minimal.Citation53 Expression of other T1D protective genetic loci Idd3 and/or Idd5 from C57BL/6 mice in NOD miceCitation54,Citation55 did not lead to changes in gut microbiota but enhanced IL-2 production and Treg function. Mullaney and co-authors further assessed the microbiota from healthy humans carrying the Idd3/5 protective alleles and found similar gut microbiota composition to NOD mice expressing the same alleles.Citation53 These studies not only support the importance of the NOD mouse model of human T1D, but also reveal the importance of the genetic susceptibility loci in T1D, which modulate the interactions of immune cells and gut microbiota.

Alterations in the gut microbiota have also been observed in humans with T1D. A study by Giongo and colleagues analyzed bacteria in fecal samples of infants and young children and discovered that children who developed T1D had higher proportions of bacteria from the Firmicutes phylum and lower proportions of bacteria in the Bacteroidetes phylum than age-matched healthy controls at 4–8 months of age.Citation37 However, by the age of 2, children who had developed T1D had a higher proportion of Bacteroidetes and a lower proportion of Firmicutes relative to healthy controls.Citation56 Rather than using stool samples, one study compared the duodenal gut microbiota of patients with T1D, or those with celiac disease (CD) to that of the healthy control subjectsCitation32 (due to proximity of the duodenum and close relationship to the pancreas). Some patients with T1D in the study had a gastroduodenal endoscopy and biopsy for CD diagnostic purposes. However, the authors found a distinctive inflammatory profile in the patients with T1D.Citation32 Patients with T1D showed overexpression of ten inflammation-associated genes in the biopsies, including chemokines and TNFα, compared to both healthy controls and CD patients.Citation32 Further, only patients with T1D exhibited an increased Firmicutes and Firmicutes/Bacteroidetes ratio but reduced proportion of Proteobacteria compared to either patients with CD or healthy controls.Citation32

In addition, some studies have investigated the gut microbiota in individuals who are positive for islet autoantibodies and those who are not. In a US-based study, the gut microbiota composition was found to be different between seropositive individuals and their seronegative first-degree relatives (FDRs) with an increased abundance of Catenibacterium, Prevotellaceae and RC9 gut group bacteria in the former.Citation57 Interestingly, the authors also found that the overall composition of gut microbiota in autoantibody-positive individuals and seronegative FDRs were similar but different from those recent-onset T1D patients and unrelated healthy controls.Citation57 It is not clear if the FDRs were living in the same or a similar environment; however, this suggests some genetic influence in the composition of gut microbiota and changes in the gut microbiota prior to and/or soon after T1D development. In a European study, Endesfelder and coauthors also compared the compostion of gut microbiota between seropositive or seronegative individuals who have an FDR with T1D.Citation58 Their results did not reveal any differences between autoantibody-positive and -negative individuals in microbiota diversity and composition, as well as single-genus abundance.Citation58 However, the authors found substantial changes in microbial interaction networks, especially in young children who later developed autoantibodies.Citation58 In another study, the microbiota composition and alpha diversity in European children, who had seroconverted and later developed diabetes, was different to those who did not seroconvert.Citation56 In addition, the children who seroconverted but had not developed diabetes by 3 years of age, had a microbiota composition and alpha diversity more similar to non-seroconverters. Together, the data confirmed microbial changes prior to and post-diabetes development. While most studies have focused on 16S rRNA sequencing of the microbiota, Pinto and colleagues investigated the microbial proteome, i.e. the proteins expressed by microbiota isolated from stool samples of healthy children and children with T1D.Citation59 The authors found that children with T1D had a higher abundance of proteins from Clostridia and Bacteroidetes, while healthy children had a higher proportion of proteins from Bifidobacterium. Although many studies provide evidence of altered gut microbiota in individuals with T1D compared to healthy control subjects, few have shown a causal link between the altered gut microbiota and the disease. However, it is important to note that some of the human studies had very small group sizes, with as few as 3–4 individuals/group.Citation37,Citation59 Thus, larger studies are needed. However, the data from the current human studies suggest that the altered microbiota and their interactions with the immune system are likely to contribute to T1D susceptibility. Therefore, it is important to do functional studies in vivo and animal models can provide the perfect tools for this purpose.

Pattern recognition receptors

Pattern recognition receptors (PRRs) are germ-line encoded and thus have been conserved over thousands of years to bind conserved structures from pathogens, designated as pathogen-associated molecular patterns (PAMPs), which are present in microorganisms.Citation60 An example of a PAMP is lipopolysaccharide (LPS), a major component of the outer membrane of gram-negative bacteria.Citation61 There are several PRR families including Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-like receptors (NLRs).

TLRs

At least 10 and 13 members of the TLR family have been identified in human and mouse, respectively, all of which recognize different PAMPs.Citation62 TLRs are expressed either on the cell surface (TLR1, 2, 4, 5, 6, 10, 11 and 12) or within the cells (TLR3, 7, 8, 9 and 13). TLRs function via signaling through the activation of the NF-κB signaling pathway, inducing proinflammatory cytokines that enable the innate immune system to quickly eradicate potential microbial threats.

There are two major TLR signaling pathways mediated by different adaptor proteins, the Myeloid differentiation primary response gene 88 (MyD88) and the Tir-domain-containing adaptor-inducing interferon β protein (TRIF). TLR3, a receptor for double stranded RNA, relies solely on TRIF signaling, while TLR4, a receptor for LPS, can signal through both MyD88 and TRIF; all the other TLRs rely on MyD88.Citation63-Citation65 Antigen presenting cells (APCs) express many TLRs and play an important role in linking gut microbiota and the host immune system.Citation66 TLR signaling is essential because it enables optimal antigen presentation by inducing APC maturation and costimulation, as well as the release of cytokines.Citation61,Citation65,Citation67Citation69

TLR interactions with the gut microbiota have been found to be important in contributing to T1D susceptibility. MyD88-deficient (MyD88−/) NOD mice are completely protected from diabetes under normal, specific-pathogen free (SPF) conditions, where gut microbiota are present.Citation70 However, diabetes was partially restored in the MyD88−/−NOD mice after antibiotic administration, suggestive of microbial involvement in the protection of the mice. Interestingly, re-deriving the MyD88−/−NOD mice to germ-free (GF) conditions abolished the disease protection. Introduction of commensal bacteria to the GF MyD88−/−NOD mice markedly reduced diabetes development. These results demonstrate that MyD88-dependent signaling is important for T1D development, which can be modulated by gut microbiota. Burrows and colleagues reported that diabetes susceptibility in TLR4−/−NOD mice and protection in TLR2−/−NOD mice were also modulated by gut microbiota.Citation71 LPS recognition has recently drawn more attention in the T1D research field. It is known that Finland has the highest T1D prevalence worldwide, whereas the incidence of T1D in Russia Karelia, a close neighbor of Finland, is six times lower.Citation72 In addition, there is a greatly reduced risk of developing other autoimmune and allergic diseases in Russion Karelia.Citation73,Citation74 Investigation of the microbiota composition in children living in the different regions revealed that Finnish children had more Bacteroides species encoding more LPS synthesis genes, when compared to Russian infants.Citation21 Furthermore, the LPS from the Bacteroides species isolated from Finnish children was structurally and functionally different from the LPS of E.coli, a Bacteroides species, found in Russian infants. When the immune function of the two different LPS types in NOD mice was tested, the LPS from Bacteroides isolate of the Finnish children was more immunostimulatory than the LPS from the E.coli isolate from the Russian children. The finding suggests that altered LPS recognition by TLR4 may be important in modulating susceptibility to T1D. Interestingly, TLR3-deficiency on the NOD background had no impact on diabetes development; however, viral infection models of diabetes development required TLR3 on both the NOD background as well as other genetic backgrounds for diabetes to develop.Citation75-Citation79 Furthermore, it was noted that enhanced costimulatory molecule expression in the islets, using transgenic constructs directed by the rat insulin promoter, involved changes to the gut microbiota and signaling through TLR3 and MyD88 pathways.Citation79 Our studies also showed that TLR9-deficient NOD miceCitation80 and TRIF-deficient NOD miceCitation81 are protected from T1D development. LPS is the ligand of TLR4 and TRIF is one of the two downstream signaling pathways of TLR4. It is interesting that diabetes protection in TRIF-deficient NOD mice is mediated by gut microbiota.Citation81 Taken together, most TLRs are required for T1D development in NOD mice, while TLR4 signaling regulates the development of T1D through gut microbiota and/or LPS.

NLRs

Another family of PRR is the nucleotide-binding oligomerization domain-like receptors (NLRs). These NLRs recognize both PAMPs and damage-associated molecular patterns (DAMPs; molecules produced by stressed cells to promote an inflammatory response). One of the best-studied NLRs is the nucleotide-binding oligomerization domain-containing protein 2 (Nod2). Mutations in this receptor mediate susceptibility to inflammatory bowel disease in humans.Citation82,Citation83 We recently showed that Nod2 also influences T1D development in NOD mice, mediated by altered gut microbiota.Citation84 In this study, Nod2−/−NOD mice were protected from T1D only when housed with other Nod2−/−NOD mice. If the Nod2−/−NOD mice were housed with Nod2-sufficient wild-type NOD mice, the Nod2−/−NOD mice developed a similar T1D incidence to WT NOD mice. This provides important evidence that the environmental conditions e.g. housing status can alter the interpretation of the disease phenotype in genetically modified mouse strains in T1D studies.

The nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome is another protein involved in the detection of pathogens by innate immune cells. We found reduced expression of chemokines and chemokine receptors on both pancreatic islets and T cells in NLRP3−/−NOD mice, which protected the mice from developing T1D.Citation85 It is currently unknown whether this process is modulated by the gut microbiota. The role of other NLR members in mediating T1D susceptibility is also unknown.

APC and gut microbiota interactions

Dendritic cells (DCs)

DCs are potent APCs found in both lymphoid and non-lymphoid tissue. Studies have shown that DCs are enriched at mucosal sitesCitation86,Citation87 and can sample microbial antigens from the gut lumen, presenting the antigens to the T cells and possibly B cells, residing in the mucosal tissue.Citation88,Citation89 Mucosal DCs express a number of chemokine receptors and tissue homing adhesion molecules including CD103, all of which facilitate the intestinal DCs to migrate to other tissue sites after antigen uptake from the gut lumen.Citation90,Citation91 DCs express a range of PRRs, and signaling through these induces maturation, promotes survival and maintains homeostasis within the DCs.Citation69,Citation92Citation98 Thus, DCs are a pivotal component of the immune system, linking innate and adaptive immunity and inducing a specific immune response to pathogens but also to self-antigens in the context of autoimmunity.

In T1D, DCs present self-antigen and activate autoreactive T cells which damage and destroy pancreatic β-cells.Citation99 DC activation through TLR9 and/or TLR3 has been shown to be important in enhancing IFNα secretion and thus promoting T cell activation and T1D development in NOD mice and humans.Citation100,Citation101 Turley and colleagues have also shown that pancreatic draining lymph nodes (PLNs) function as an intersection for immune responses to gut non-self antigen and pancreatic self-antigen.Citation102 It is conceivable that some microbial antigens may share antigenic homology with pancreatic self-antigen, which can be sampled by intestinal DCs and promote undesirable autoimmune responses. In this regard, we have recently shown that autoreactive and diabetogenic CD8 T cells recognize a microbial peptide and promote the acceleration of T1D development in a T cell receptor (TCR) transgenic NOD mouse.Citation103

B cells

B cells originate and develop in the bone marrow. Mature B cells can differentiate to antibody-secreting plasma cells. IgM is the first antibody that B cells produce in response to pathogen invasion. However, IgM is usually low affinity and hence the need for affinity maturation including class switching, i.e., Ig gene rearrangements, to generate antibodies with high affinity, in order to more specifically target the antigen and control pathogen invasion.Citation104-Citation106 B cells undergo class switching in response to different antigens. The type of class-switched antibody produced by B cells is also determined by the tissue location. For example, IgA is the predominant antibody in the mucosal tissues, while IgG is the most prevalent antibody in the circulation.Citation107 B cells often require T cell help to complete class switching; however, commensal bacteria and/or bacterial antigens presented by mucosal DCs can induce B cells to secrete IgA, which protects the mucosa from invasion by pathogens and reduces inflammatory signals.Citation108,Citation109 Intestinal epithelial cells can also induce IgA secretion.Citation110

In addition to antibody secretion, B cells also function as antigen presenting cells, promoting diabetes development.Citation111-Citation113 Although T1D is considered a T-cell mediated disease, it has become clear that B cells play an important role in the development of T1D. The precise role of B cells in the pathogenesis of T1D is complex and not fully understood; however, the autoantibodies produced by self-reactive B cells provide a very good biomarker to predict T1D onset in the individuals with high risk of developing T1D. The self-reactive B cells in T1D produce IgG autoantibodies to a range of antigens including insulin,Citation114 GAD,Citation115 IA-2Citation116 and ZnT8.Citation117 Less is known about the role of IgA in T1D, although data from two T1D studies provide conflicting evidence as to whether the IgA concentrations in the serum are different between controls and T1D patients.Citation118,Citation119 IgA is enriched in mucosal sites of both mouse and human in a secretory form and it is estimated that approximately 40 mg/kg body weight of IgA are produced in human intestine.Citation120,Citation121 Detection of immunoglobulins bound to the bacteria within the intestineCitation122,Citation123 may provide new avenues of investigation for T1D studies, enabling us to understand which gut microbiota become antibody targets and how the immune system interacts with them.

B cells in NOD mice express enhanced levels of some PRRs (and associated molecules) compared to C57BL/6 mice.Citation124 Further, unmethylated CpG deoxyoligonucleotides, which are a ligand for TLR9, activated pro-B cells. These cells have a regulatory function and are capable of protecting NOD mice from T1D development.Citation125 Interestingly, the C1858T (lyp) polymorphism of the PTPN22 gene in humans was also associated with altered B cell responses in individuals with T1D after stimulation with CpG.Citation126 Therefore, innate signals can mediate important changes altering susceptibility to T1D in both NOD mice and humans. However, B cells are not the only adaptive immune cells to be influenced by the gut microbiota.

T cells are influenced by gut microbiota

T cells are a major player in the adaptive immune responses that both fight against pathogens and regulate immune responses to maintain immuno-homeostasis. T cells originate from bone marrow stem cells and undergo development in the thymus (including the gene rearrangements that enable the antigen-specific TCR to be expressed). Thymic T cells also undergo selection processes, ensuring the deletion of highly auto-reactive T cells, which prevents autoimmunity. However, this process is not complete and may contribute to the development of autoimmune disorders in people who are susceptible to autoimmunity.

T cells expressing their antigen-specific TCR, can recognize a vast diversity of antigens, pathogens and non-pathogens, which include self-antigens.Citation127 However, the antigens recognized by the TCR are presented by APCs through the antigenic peptide-MHC complex. The APCs express costimulatory molecules and produce cytokines, and these, together with the recognition of specific antigen, stimulate T cells to differentiate into different effector subsets. Studies have shown that commensal bacteria, most likely mediated by APCs, can induce T helper (Th) 1,Citation128,Citation129 Th2,Citation130,Citation131 Th17Citation132,Citation133 and T follicular helper (Tfh) cells.Citation134 For example, the presence of Segmented Filamentous bacteria (SFB) in the gut can induce IL17-producing CD4+ Th17 cells,Citation132,Citation133 and Tfh cells.Citation134 Targeting SFB specifically may enable potential treatments for Th17-driven autoimmune diseases e.g. Experimental Autoimmune Encephalomyelitis (EAE, a mouse model of multiple sclerosis) and collagen-induced arthritis in mice.Citation135,Citation136 Although, T1D is driven by Th1 cells, SFB have been shown to protect NOD mice from T1D when housed in SPF conditions.Citation137 However, SFB introduction into GF NOD mice had no effect on T1D development in female mice, whereas there was a significant delay in T1D onset in male GF mice.Citation138 Other studies have shown the inter-regulatory relationship between Th1 and Th17, where Th17 cells are also controlled by Treg cells. It is plausible that the Th1/Th17/Treg axis plays an important role in modulating T1D susceptibility by alteration of gut microbiota. The newly-identified role of SFB in altering Tfh differentiation and trafficking in a mouse model of arthritis is interesting.Citation134 This provides evidence of the microbiota altering T:B cell interactions within the germinal centers prior to autoimmunity development. In T1D, Tfh T cells from diabetic NOD mice can transfer diabetesCitation139 and Tfh cells were found to be increased in T1D patients.Citation140 A recent study investigating insulin-specific T:B cell interactions in NOD mice, also revealed an increase in Tfh cells associated with increased diabetes development.Citation141 Therefore further understanding antigen-specific germinal center interactions and the influence of gut microbiota, prior to autoimmunity development may be very important.

Treg cells

Tregs are characterized by their expression of the forkhead box transcription factor (FoxP3),Citation142 and are potent at suppressing immune responses.Citation143 Tregs were previously named suppressor T cells,Citation144 and we have gained much more understanding of these cells in recent years. Tregs can be generated within the thymus (natural Tregs, nTregs) or in the periphery (induced Treg, iTregs). FoxP3 deficiency, caused by the Scurfy x-linked mutation,Citation145 resulted in severe immune cell infiltration in multiple organs and autoimmune destruction.Citation146,Citation147 Thus, Treg cells are vital in mediating immune tolerance to autoantigens. In addition, they are also very important in limiting the immune response to foreign antigens to prevent tissue damage.

Tregs, like other immune cells, also express TLRs including TLR4, TLR5, TLR7 and TLR8 as identified by real time PCR.Citation148 Upon LPS stimulation, that is recognized via TLR4, Tregs became more activated and exhibit enhanced suppressive capabilities.Citation148 Studies have shown that commensal bacteria, such as strains of Clostridium, by promoting a TGF-β rich environment, induce T effectors to become Tregs in the colon and protect mice from chemical induced colitis.Citation149 Treg induction was also confirmed in mice given a mixture of human stool-isolated Clostridium species.Citation150 Interestingly, Treg induction by Clostridium species was independent of PRR signaling, as Treg induction was not impaired in a number of PRR deficient mice including MyD88−/− mice.Citation149 However, other studies have shown that Treg conversion in colonized germ-free mice with the altered Schaedler flora (ASF, a mixture of 8 strains of human gut bacteria including a Clostridium species) is MyD88 dependent.Citation151 It is not clear whether other strains of bacteria in the ASF also contribute to the Treg conversion. However, these studies suggest that Treg induction and the mechanism behind it depend on the type of bacteria. More recently, Nod2, a member of the NLR family, which recognizes the bacterial component muramyl dipeptide (MDP) was also found to regulate human Treg survival by preventing apoptosis induced by MDP stimulation; however, Tregs from patients with IBD, who have the Nod2 gene mutation, were not protected from apoptosis.Citation152 We have shown recently that Nod2-deficient NOD mice are protected from T1D development, which was, at least in part, mediated by increased gut microbiota-induced Tregs in the pancreatic lymph nodes.Citation84 This data shows the microbiota and immune recognition together shape the regulatory T cell response and may provide an important target for therapy.

Metabolism and the gut microbiota

The intestinal microbiota utilize undigested food products as substrates for fermentation resulting in the production of different metabolites. Short-chain fatty acids (SCFAs), saturated fats, L-carnitine, and choline are examples of microbially-derived metabolites. Therefore, the presence of these metabolites depends on the microbiota composition. Studies have shown that the metabolites, especially SCFAs influence the differentiation and function of immune cells; thus, they may play an important role in the development of T1D.

Other metabolic components including sex hormones can affect the immune system including the induction of autoimmunity. The female gender bias in T1D development seen in the NOD mice has been shown to be consequent upon interactions between sex hormones and gut microbiota.Citation138,Citation153 The gut microbiota in male NOD mice influence the levels of testosterone, and higher levels of testosterone are associated with the protection against T1D development.Citation153 Several human autoimmune disorders have a strong gender bias, with a higher incidence in women. While human T1D in adulthood has a small gender bias towards men, it is important to decipher the role(s) of hormones and microbiota including microbial products in mediating susceptibility to autoimmune diseases.

Metabolites

One of the most significant metabolites produced by gut microbiota are SCFAs, during the fermentation of dietary fiber in the colon. Butyrate, propionate, and acetate are the major SCFAs produced in the gut, which can regulate the host immune system, central nervous system, gastrointestinal system, and metabolism through various mechanisms. The oxidation of SCFAs, butyrate in particular, is the principal energy source utilized by colonocytes.Citation154,Citation155 Butyrate also enhances the integrity of human and mouse intestinal epithelial cells. This occurs by controlling the assembly of tight junctions as demonstrated in an in vitro culture systemCitation156 and in mice in vivo through the stabilization of the hypoxia-inducible factor (HIF), a transcription factor important for mediating epithelial barrier functions.Citation157 Moreover, butyrate can be sensed by the immune system to promote Treg induction, concomitant with decreased inflammation in the intestines.Citation158-Citation160

The short chain fatty acid propionate is converted into glucose in the intestine, resulting in decreased glucose production from the liver.Citation161 Propionate also acts as an agonist of FFAR3, inducing the peripheral nervous system to alter host metabolism by decreasing adiposity, body weight and glucose production in the liver, thus promoting better glucose control.Citation162 In addition, SCFAs can regulate the expression of peptide YY (an enteroendocrine hormone) that controls gut motility and transit rate, as well as SCFA uptake and can promote anti-inflammatory properties, offering protection from induced inflammatory diseases such as colitis, arthritis and asthma in mouse models.Citation163,Citation164 Thus, increased production of SCFAs from dietary fiber supplements or the ingestion of probiotics can inhibit pro-inflammatory cytokines and chemokines that could be used in potential treatments for autoimmunity e.g. colitis. However, a balance would be required, as SCFAs are an additional source of calories and can be associated with obesity and metabolic syndrome.Citation165

While less is known about the role of SCFAs in the development of T1D, a few recent studies have suggested that SCFAs can modulate T1D susceptibility. One study showed that feeding NOD mice with acetylated or butyrated high-amylose maize starch diets increased their serum concentrations of acetate or butyrate and protected NOD mice from developing diabetes.Citation166 Further, protection was enhanced if NOD mice were fed with a combination of both acetylated and butyrated diets. Interestingly, NOD mice fed with the high acetate diet had reduced frequencies and numbers of islet autoantigen-reactive T cells in the spleen and pancreatic lymph nodes (PLNs). However, the protection in NOD mice fed with a high butyrate-containing diet was related to increased Treg number with enhanced suppressive functions.Citation166 Another study demonstrated that butyrate could influence the secretion of cathelicidin-related antimicrobial peptide (CRAMP) by islets, with higher concentrations of CRAMP associated with protection from T1D development.Citation167 CRAMP expression was shown to alter the islet microenvironment, by inducing tolerogenic islet macrophages, regulatory DCs and Tregs, all of which facilitate the reduction of autoreactive T cell activation and thus prevent diabetes development. Altering the microbiota composition by fecal transfer or by antibiotic treatment can change the availability of metabolites, which in turn, alters the risk of developing T1D.Citation168,Citation169 Therefore, these options may prove useful in developing prevention therapies. However, the relationship between microbial metabolism and human T1D is still not well understood, and more studies in this area are needed in the future.

Diet and type 1 diabetes

Diet is well known to influence microbial composition and functions. In Burkina Faso, Africa, where diets consist of an abundance of complex carbohydrates resulting in higher microbial diversity, children produce greater amounts of SCFAs when compared to children from Europe.Citation170 In contrast, GF mice colonized with human stool bacteria from an individual with a Western-style diet had less microbial diversity and a worsened ability to metabolize complex carbohydrates. However, those mice given the stool bacteria from the individuals with an enriched microbiota-accessible carbohydrate diet had increased microbial diversity and were able to metabolize complex carbohydrates.Citation16,Citation171 Interestingly, numerous studies have suggested that Prevotella:Bacteroides ratios are related to the dietary intake of either complex carbohydrate diets or proteins and fats respectively.Citation170,Citation172 Of note, when dietary fibers are almost completely fermented, the pH of the large intestine increases, providing a reduction in butyrate-producing microbiota but an increase in acetate- and proprionate-producing Bacteroides bacteria.Citation173

A barley kernel-based bread diet was introduced to healthy subjects for 3 days in a recent dietary intervention study.Citation174 The authors identified the improvement of glucose metabolism to be associated with an increased abundance of Prevotella copri.Citation174 The presence of Prevotella in these individuals had enhanced enzymatic activity related to breaking down complex carbohydrates, which promotes the generation of a number of SCFAs. The colonization of GF mice with microbiota from the study participants also confirmed an increased abundance of Prevotella that was associated with improved glucose metabolism. Furthermore, branched-chain fatty acid produced by the fermentation of branched-chain amino acids correlated with insulin resistance in germ-free mice receiving stool bacteria from obese human individuals.Citation175

Dietary influence on T1D has been examined both as a causative agent as well as a preventative or modulating factor. Oral administration of nicotinamide, a vitamin B group substance, to NOD mice prevented the development of diabetes.Citation176 Further, NOD mice were completely protected from T1D development when administered nicotinamide in combination with a diet consisting of an infant formula, where soy was the source of protein.Citation177 Vitamin D, specifically the active form, 1,25 di-hyroxyvitamin D, has also been shown to prevent from severe insulitis due to the increased Treg cell activity.Citation178-Citation180

Gluten, a component of wheat protein, has many antigenic properties, and has been implicated in the pathogenesis of several autoimmune disease states (predominantly CD) but it has also been implicated in T1D. It has been reported that CD can affect up to ~ 10% of individuals with T1D due to the overlap in HLA-DR3/DQ2 genetic susceptibility between the two diseases.Citation181,Citation182 Furthermore, tissue transglutaminase antibodies are present in some T1D patients, who share susceptibility SNPs to CD in the CTLA4 gene.Citation183 Even in those individuals with T1D who were negative for the tissue transglutaminase autoantibody in the serum, the antibody was still found in the jejunum.Citation184 Interestingly, T1D patients have increased expression of duodenal inflammatory chemokines and cytokines compared to CD patients and healthy controls.Citation32 This was also associated with altered microbiota in those T1D patients. Gluten-free diets have been shown to protect NOD mice from the development of T1D.Citation185-Citation187 Two studies have shown that gluten in the diet affected the quantity and composition of the gut microbiota. Citation185,Citation186 Funda and colleagues also reported that while a gluten-free diet prevented diabetes in NOD mice, a gluten-enriched diet also had a preventative effect.Citation187 They hypothesized that gluten itself is not diabetogenic but can have an immunomodulatory effect on a diabetes-susceptible host by altering the gut microbiota which in turn regulates the immune system. Human studies investigating how diet may influence T1D susceptibility are currently underway. The BABYDIET study is a prospective primary prevention trial recruiting children “at risk” of developing diabetes, who have a first-degree relative with T1D and carry a T1D-risk HLA genotype. Children were randomly assigned to one of two groups whereby gluten was introduced in the diet at 6 months or 1 year of age.Citation188 Data from the participants within the first 3 years of age showed similar prevalence of islet autoimmunity regardless of when gluten was introduced; however, these children will continue to be followed over time, to observe generation of autoantibodies and other markers that are associated with the development of both celiac and T1D autoimmunity.

Antibiotic usage and type 1 diabetes

There are many different classes of antibiotics that target different bacterial pathogens. In addition to clearing the pathogen(s) from the original infection in various body sites, antibiotics can alter the gut microbiota directly, particularly as most of the antibiotics are taken via oral route. Will antibiotic usage affect T1D development by changing gut microbiota? Studies in mouse models of T1D suggest that the type of antibiotics, time of usage (early in life or later in life) and the duration of usage can modify the susceptibility of T1D development. summarizes different studies conducted to assess the role of microbial modulation in the development of T1D in NOD mice. We have shown that vancomycin, which predominantly targets Gram-positive bacteria, promoted T1D development in NOD mice, whereas neomycin, which targets Gram-negative bacteria, protected NOD mice from T1D development.Citation189,Citation193 Interestingly, others have reported that life-long vancomycin usage protected NOD mice from T1D development.Citation191 It is clear that not all antibiotics have the same impact on T1D development. Some antibiotics can protect NOD mice from the development of T1D,Citation189,Citation191,Citation193 while others promote the development of diabetes in NOD mice.Citation168,Citation190,Citation192 Interestingly, we found that the treatment of combination of four different antibiotics (ampicillin, vancomycin, metronidazole and neomycin, AVMN) had no effect on diabetes development in NOD mice despite the fact that the AVMN treatment depleted most, if not all, gut bacteria (Peng, et al., unpublished). This highlights the observation that the treatment protocol can also influence diabetes development. In the study showing that vancomycin protected NOD mice, the mice were treated from birth to 28 days of age or from 8 weeks of age to termination.Citation191 However, in the studies showing that vancomycin accelerated T1D development, NOD mice were treated from conception to termination or from conception to 24 hours post-birth.Citation189,Citation190 The different treatment protocols, including dose, duration and starting age for these antibiotic studies make it difficult to compare the outcome of different studies directly. Moreover, the gender of the mice used in the studies may also affect diabetes development. As discussed earlier, sex hormones influence the gut microbiota and T1D susceptibility in the NOD mouse.Citation138,Citation153 While most studies have used female mice, some studies have been conducted in both genders. Candon and co-authors found that long-term (from conception to the progeny at 40 wks of age) treatment with vancomycin or a combination of streptomycin, colistin and ampicillin (Strep-Col-Amp) significantly increased diabetes development in male NOD mice but had no effect on female mice.Citation190 In another study, male NOD mice receiving tylosin also exhibited increased diabetes development.Citation168 Interestingly, the female NOD mice, also exhibited increased diabetes development in one animal facility but this was not reproducible in a second animal facility. This raises an important point that the resident gut microbiota and animal facility standards can influence the experimental outcome. This also raises the issue of reproducibility of studies as small microbial changes may influence the effectiveness of the antibiotic treatment. Clearly, these studies are not possible in humans; however, epidemiological investigations into antibiotic usage and T1D development in humans have not shown any evidence that antibiotics administered to children have influenced the onset of T1D.

Table 1. Antibiotic Treatment effects on diabetes development in NOD mice.

Future directions: manipulating the gut microbiota as a novel therapy

Probiotics

Probiotics are microorganisms that may have health benefits by modifying the gut microbiota and improve nutrient absorption, enhance immune regulation and protect the host from infection and disease.Citation194 While many of the probiotics may not colonize their hosts in the long-term, they do have important immunomodulatory effects in the short-term.Citation195

Probiotics have been shown to protect NOD mice and BB rats from developing T1D.Citation196-Citation199 Lactobacillus casei, which is believed to be a probiotic strain, can protect NOD mice from T1D development when administered in the diet from 4 weeks of age.Citation196 This protection was associated with decreased splenic CD8 T cell number and increased IL-10 and IL-2 cytokines with age. The probiotic VSL#3 is a mixture of bifidobacteria (B. longum, B. infantis and B. breve), lactobacilli (L. acidophilus, L.casei, L. delbrueckii subsp. L. bulgaricus and L. plantarum) and a strain of streptococcus bacteria (Streptococcus salivarius subsp. thermophilus).Citation197 In a study, Calcinaro and colleagues administered VSL#3 to NOD mice, three times per week from 4 to 32 weeks of age and this protocol led to significant protection from T1D development in treated NOD mice compared to non-treated controls (21% vs 81% respectively). VSL#3 administration decreased expression of IL-1β and increased expression of indoleamine 2,3-dioxygenase and IL-33, both of which have tolerogenic properties.Citation199 However, in another study, VSL#3 was administered via the drinking water to pregnant mice, just before birth until termination of the progeny (22 wks old), and this did not protect NOD mice from the development of T1D.Citation169 In a very recent study, Hanninen and colleagues reported that Akkermansia muciniphila abundancy is negatively correlated to T1D development and oral transfer of A. muciniphila delays diabetes development in NOD/Jax mice that, otherwise, have an early disease onset.Citation200 However, A. muciniphila did not reduce the overall incidence of diabetes,Citation200 whereas gavage of Clostridium butyricum CGMCC0313.1 was able to significantly reduce the incidence of diabetes in NOD mice by promoting regulatory T cells.Citation201

There have been some studies investigating probiotics and host immunity in humans. Treating CD children with two Bifidobacterium breve strains, Primec and co-authors identified changes in microbiota composition that were associated with the changes in SCFAs and reduced TNFα in circulation.Citation202 The authors also found that Verrucomicrobia, Parcibacteria and some other bacteria yet unknown phyla were strongly correlated to TNFα.Citation202 The probiotic administration to the CD children lowered the level of TNFα and reduced the abundance of Verrucomicrobia.Citation202 This may provide potential therapeutic targets for CD. However, in a different disease setting, a reduced proportion of Verrucomicrobia has also been associated with glucose intolerance in type 2 diabetes patients and prediabetes subjects.Citation203 There have also been a few studies in T1D. A double-blind randomized pilot study investigated how Lactobacillus johnsonii N6.2, which was shown to delay T1D onset in BB rats,Citation198 affects the host immunity in healthy adults.Citation204 In this phase I human study, only healthy individuals without gastrointestinal disorders or other health issues (e.g. diabetes, mental diseases, kidney and heart diseases and others) were recruited for the study. The administration of L. johnsonii N6.2 (taken in capsules for over 8 weeks) resulted in an increase of tryptophan in the circulation.Citation204 It is known that tryptophan promotes Treg cell induction and expansion as well as suppresses the differentiation of Th1 cells. These data suggest that L. johnsonii N6.2 is safe for treatment in adults, although the authors found that the study subjects had increased monocytes, NK cells and CD8 T cells in the peripheral blood compared to the placebo controls.Citation204 It is clear that more studies need to be done before launching clinical trials in the subjects with T1D or those at risk of developing T1D.

The Environmental Determinants of Diabetes in the Young (TEDDY) study follows children at risk of developing T1D to understand how different environmental factors may influence T1D susceptibility in humans. Recent data from the TEDDY study have revealed a large variability in the fecal microbial probiotic (total lactobacilli and L. plantarum) compositions between the children, particularly before 10 months of age.Citation205 It is currently unknown whether those with a reduced abundance of lactobacilli in early life will have a greater risk of developing T1D; however, the TEDDY study shows the presence of probiotic-related bacteria in children who are at risk of developing T1D, and correlating this with development of diabetes, over time, will be of considerable interest. This is an ever-expanding area with more probiotic-based human T1D clinical trials planned.

Fecal microbiota transplantation

Fecal microbiota transplantation (FMT) can change the gut microbiota in the recipients to elicit health benefits. FMT involves processing stool from a healthy donor (for allogeneic transplants) or from the recipient (autologous transplant), although more often the FMT is an allogeneic transplant from a healthy donor. Sometimes the FMT recipients may need to be treated with antibiotics 24–72 hours prior to FMT to eliminate any existing deleterious gut microbiota. An alternative approach to fast, and have bowel preparaton shortly before the treatment. FMT has been used for the treatment of severe Clostridium difficile-induced colitis.Citation206 Many studies confirmed that FMT therapy is safe and effective in treating human IBD.Citation207 Interestingly, FMT (from lean donor feces) improves insulin sensitivity in individuals with metabolic syndrome.Citation208

In T1D, promising results using FMT have been shown in mice. Our study showed that fecal materials from MyD88−/−NOD mice, which were protected from diabetes, can delay and reduce diabetes development in NOD mouse recipientsCitation209 However, there has been no report to date on whether FMT could be beneficial in ameliorating disease in T1D patients or in preventing T1D development in the individuals who are at risk of developing T1D. Both NOD mouse studies and recent TEDDY studies have pointed to early life as an important window of opportunity for effective intervention to reduce islet autoimmunity, especially in relation to the composition of gut microbiotaCitation56 and possibly FMT.

Concluding remarks

Increasing evidence suggests that there are microbial perturbations in individuals with islet autoimmunity or T1D compared to healthy control subjects; however, we are still not clear about the defined mechanisms. Therefore, further functional studies are needed, not only to probe the interaction of microbiota with immune system but also to identify the causal link(s) between the presence of certain gut bacteria and diabetogenic autoimmune responses. Germ-free NOD mice will provide a valuable tool in deciphering the role of the microbiota in relation to the development of T1D, both in vitro, and more importantly, in vivo. While GF mice are known to have a less mature immune system, particularly in relation to the gut-associated mucosal lymphoid tissue,Citation210 GF NOD mice, still develop T cell-mediated T1D.Citation71,Citation138,Citation153,Citation211 Thus, using these GF NOD mice as recipients for human stool from diabetic donors and healthy control donors, allows us to understand how the microbiota may modulate the immune system in T1D in vivo. Studies in obesity and T2D have shown that human gut microbiota can improve glucose control and insulin resistance and thus manipulation of the microbiota may also be used to achieve better glycemic control in patients with T1D.

Diet and probiotics may provide easier acceptance and compliance in participants for disease prevention and/or intervention and/or modulation of disease. However, there are still some challenges e.g. in dietary interventions, the subjects have to be at the age of taking solid food. As for the use of probiotics, the question remains as to how long the beneficial immunomodulatory effects induced by probiotic will last and if further doses are required. Moreover, while FMT may alter the gut microbiota in the longer term, many of these studies pre-treat with antibiotics which provides a niche allowing newly introduced microbiota to colonize. It is noteworthy that antibiotic treatment may lead to antibiotic resistance. In addition, it is highly possible that FMT-induced microbiota changes can be modified by diet and other factors. Therefore, for future therapy development, we require further understanding of 1) microbial community interactions as a complex ecosystem; 2) the interaction of host microbiome with intestinal micro-environment including gut epithelial cells and specialized gut endocrine cells including Goblet cells and Paneth cells and 3) the interaction of gut microbiota with the host immune cells locally and systemically. Furthermore, we also need to better define subgroups of patients, as increasing evidence suggests that T1D is not a homogenous disease condition. If we can combine the information on microbiota with clinical data, to truly decipher the results by subgrouping patients based on resident microbiota prevalence, c-peptide concentrations, autoantibody presence and type, as well as autoreactive T cell information, we will be able to design treatment with more precision. In summary, the role of microbiota in T1D is complex. While there are associations with microbial composition changes at the time of seroconversion and changes associated with disease onset, it remains unclear if the microbiota play a causal role in human T1D development. However, studies in NOD mice have shown that microbial antigens stimulated diabetogenic CD8 T cells and accelerated T1D development.Citation103,Citation212 It is clear from mouse and human studies, the microbiota can induce both proinflammatory and anti-inflammatory changes of the immune system.

Disclosure of potential conflicts of interest

No potential conflicts of interest

Additional information

Funding

This work was supported by a JDRF postdoctoral fellowship to JAP (3-PDF-2016-197-A-N), a Discovery to Cure High School Internship to AA and research grants from NIH (DK092882, DK100500, and P30 DK945735) and ADA (1-14-BS-222) to LW.

References

  • Hollister EB, Riehle K, Luna RA, Weidler EM, Rubio-Gonzales M, Mistretta TA, Raza S, Doddapaneni HV, Metcalf GA, Muzny DM, et al. Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome. 2015;3:36. doi:10.1186/s40168-015-0101-x.
  • Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: the unseen majority. Proc Natl Acad Sci U S A. 1998;95:6578–6583.
  • Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi:10.1038/nature08821.
  • Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, Arumugam M, Kultima JR, Prifti E, Nielsen T, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–841. doi:10.1038/nbt.2942.
  • Sender R, Fuchs S, Milo R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016;14:e1002533. doi:10.1371/journal.pbio.1002533.
  • Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009;326:1694–1697. doi:10.1126/science.1177486.
  • Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148:1258–1270. doi:10.1016/j.cell.2012.01.035.
  • Oh KJ, Lee SE, Jung H, Kim G, Romero R, Yoon BH. Detection of ureaplasmas by the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. J Perinat Med. 2010;38:261–268. doi:10.1515/JPM.2010.040.
  • Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48:8–12. doi:10.1111/j.1472-765X.2008.02475.x.
  • Hu J, Nomura Y, Bashir A, Fernandez-Hernandez H, Itzkowitz S, Pei Z, Stone J, Loudon H, Peter I, Tse H. Diversified microbiota of meconium is affected by maternal diabetes status. PLoS One. 2013;8:e78257. doi:10.1371/journal.pone.0078257.
  • Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, Narbad A, Olivares M, Xaus J, Rodríguez JM. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol. 2005;51:270–274. doi:10.1007/s00284-005-0020-3.
  • Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, Björkstén B, Engstrand L, Andersson AF. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut. 2014;63:559–566. doi:10.1136/gutjnl-2012-303249.
  • Huh SY, Rifas-Shiman SL, Zera CA, Edwards JW, Oken E, Weiss ST, Gillman MW. Delivery by caesarean section and risk of obesity in preschool age children: a prospective cohort study. Arch Dis Child. 2012;97:610–616. doi:10.1136/archdischild-2011-301141.
  • Khashan AS, Kenny LC, Lundholm C, Kearney PM, Gong T, Almqvist C. Mode of obstetrical delivery and type 1 diabetes: a sibling design study. Pediatrics. 2014;134:e806–13. doi:10.1542/peds.2014-0819.
  • Cheng J, Ringel-Kulka T, Heikamp-De Jong I, Ringel Y, Carroll I, De Vos WM, Salojärvi J, Satokari R. Discordant temporal development of bacterial phyla and the emergence of core in the fecal microbiota of young children. ISME J. 2016;10:1002–1014. doi:10.1038/ismej.2015.177.
  • Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64. doi:10.1038/nature18846.
  • Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535:75–84. doi:10.1038/nature18848.
  • Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015;125:926–938. doi:10.1172/JCI76304.
  • Mayer-Davis EJ, Dabelea D, Lawrence JM. Incidence trends of type 1 and type 2 diabetes among youths, 2002-2012. N Engl J Med. 2017;377:301. doi:10.1056/NEJMc1706291.
  • Patterson CC, Gyurus E, Rosenbauer J, Cinek O, Neu A, Schober E, Parslow RC, Joner G, Svensson J, Castell C, et al. Trends in childhood type 1 diabetes incidence in Europe during 1989-2008: evidence of non-uniformity over time in rates of increase. Diabetologia. 2012;55:2142–2147. doi:10.1007/s00125-012-2571-8.
  • Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD, Hämäläinen A-M, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165:842–853. doi:10.1016/j.cell.2016.04.007.
  • Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y. Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu. 1980;29:1–13.
  • Nakhooda AF, Like AA, Chappel CI, Murray FT, Marliss EB. The spontaneously diabetic Wistar rat. Metabolic and morphologic studies. Diabetes. 1977;26:100–112.
  • Anderson MS, Bluestone JA. The NOD mouse: a model of immune dysregulation. Annu Rev Immunol. 2005;23:447–485. doi:10.1146/annurev.immunol.23.021704.115643.
  • Bortell R, Yang C. The BB rat as a model of human type 1 diabetes. Methods Mol Biol. 2012;933:31–44. doi:10.1007/978-1-62703-068-7_3.
  • Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464:1293–1300.
  • Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155:173–181. doi:10.1111/j.1365-2249.2008.03860.x.
  • Voorbij HA, Jeucken PH, Kabel PJ, De Haan M, Drexhage HA. Dendritic cells and scavenger macrophages in pancreatic islets of prediabetic BB rats. Diabetes. 1989;38:1623–1629.
  • Miyazaki A, Hanafusa T, Yamada K, Miyagawa J, Fujino-Kurihara H, Nakajima H, Nonaka K, Tarui S. Predominance of T lymphocytes in pancreatic islets and spleen of pre-diabetic non-obese diabetic (NOD) mice: a longitudinal study. Clin Exp Immunol. 1985;60:622–630.
  • Zhang L, Nakayama M, Eisenbarth GS. Insulin as an autoantigen in NOD/human diabetes. Curr Opin Immunol. 2008;20:111–118. doi:10.1016/j.coi.2007.11.005.
  • Gale EA, Gillespie KM. Diabetes and gender. Diabetologia. 2001;44:3–15. doi:10.1007/s001250051573.
  • Pellegrini S, Sordi V, Bolla AM, Saita D, Ferrarese R, Canducci F, Clementi M, Invernizzi F, Mariani A, Bonfanti R, et al. Duodenal mucosa of patients with type 1 diabetes shows distinctive inflammatory profile and microbiota. J Clin Endocrinol Metab. 2017;102:1468–1477. doi:10.1210/jc.2016-3222.
  • Vaarala O, Atkinson MA, Neu J. The “perfect storm” for type 1 diabetes: the complex interplay between intestinal microbiota, gut permeability, and mucosal immunity. Diabetes. 2008;57:2555–2562. doi:10.2337/db08-0331.
  • Daft JG, Lorenz RG. Role of the gastrointestinal ecosystem in the development of type 1 diabetes. Pediatr Diabetes. 2015;16:407–418. doi:10.1111/pedi.12282.
  • Kuitunen M, Saukkonen T, Ilonen J, Akerblom HK, Savilahti E. Intestinal permeability to mannitol and lactulose in children with type 1 diabetes with the HLA-DQB1*02 allele. Autoimmunity. 2002;35:365–368.
  • Sapone A, de Magistris L, Pietzak M, Clemente MG, Tripathi A, Cucca F, Lampis R, Kryszak D, Cartenì M, Generoso M, et al. Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes. 2006;55:1443–1449.
  • Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyöty H, et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 2011;5:82–91. doi:10.1038/ismej.2010.92.
  • Nguyen TL, Vieira-Silva S, Liston A, Raes J. How informative is the mouse for human gut microbiota research? Dis Model Mech. 2015;8:1–16. doi:10.1242/dmm.017400.
  • Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989;299:1259–1260.
  • Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002;347:911–920. doi:10.1056/NEJMra020100.
  • Martins TC, Aguas AP. Mechanisms of Mycobacterium avium-induced resistance against insulin-dependent diabetes mellitus (IDDM) in non-obese diabetic (NOD) mice: role of Fas and Th1 cells. Clin Exp Immunol. 1999;115:248–254.
  • Zaccone P, Raine T, Sidobre S, Kronenberg M, Mastroeni P, Cooke A. Salmonella typhimurium infection halts development of type 1 diabetes in NOD mice. Eur J Immunol. 2004;34:3246–3256. doi:10.1002/eji.200425285.
  • Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U, Shaw JE. Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 2014;103:137–149. doi:10.1016/j.diabres.2013.11.002.
  • Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282:20143085. doi:10.1098/rspb.2014.3085.
  • Roesch LF, Lorca GL, Casella G, Giongo A, Naranjo A, Pionzio AM, Li N, Mai V, Wasserfall CH, Schatz D, et al. Culture-independent identification of gut bacteria correlated with the onset of diabetes in a rat model. ISME J. 2009;3:536–548. doi:10.1038/ismej.2009.5.
  • Daft JG, Ptacek T, Kumar R, Morrow C, Lorenz RG. Cross-fostering immediately after birth induces a permanent microbiota shift that is shaped by the nursing mother. Microbiome. 2015;3:17. doi:10.1186/s40168-015-0080-y.
  • Wen L, Wong FS, Burkly L, Altieri M, Mamalaki C, Kioussis D, Flavell RA, Sherwin RS. Induction of insulitis by glutamic acid decarboxylase peptide-specific and HLA-DQ8-restricted CD4(+) T cells from human DQ transgenic mice. J Clin Invest. 1998;102:947–957. doi:10.1172/JCI2723.
  • Wen L, Wong FS, Tang J, Chen NY, Altieri M, David C, Flavell R, Sherwin R. In vivo evidence for the contribution of human histocompatibility leukocyte antigen (HLA)-DQ molecules to the development of diabetes. J Exp Med. 2000;191:97–104.
  • Wen L, Chen NY, Tang J, Sherwin R, Wong FS. The regulatory role of DR4 in a spontaneous diabetes DQ8 transgenic model. J Clin Invest. 2001;107:871–880. doi:10.1172/JCI11708.
  • Nishimoto H, Kikutani H, Yamamura K, Kishimoto T. Prevention of autoimmune insulitis by expression of I-E molecules in NOD mice. Nature. 1987;328:432–434. doi:10.1038/328432a0.
  • Wicker LS, Appel MC, Dotta F, Pressey A, Miller BJ, DeLarato NH, Fischer PA, Boltz RC, Peterson LB. Autoimmune syndromes in major histocompatibility complex (MHC) congenic strains of nonobese diabetic (NOD) mice. The NOD MHC is dominant for insulitis and cyclophosphamide-induced diabetes. J Exp Med. 1992;176:67–77.
  • Silverman M, Kua L, Tanca A, Pala M, Palomba A, Tanes C, Bittinger K, Uzzau S, Benoist C, Mathis D. Protective major histocompatibility complex allele prevents type 1 diabetes by shaping the intestinal microbiota early in ontogeny. Proc Natl Acad Sci U S A. 2017;114:9671–9676. doi:10.1073/pnas.1712280114.
  • Mullaney JA, Stephens JE, Costello ME, Fong C, Geeling BE, Gavin PG, Labrecque M, Joly S, Yergeau E, Brereton NJB. Type 1 diabetes susceptibility alleles are associated with distinct alterations in the gut microbiota. Microbiome. 2018;6:35. doi:10.1186/s40168-018-0432-5.
  • Yamanouchi J, Rainbow D, Serra P, Howlett S, Hunter K, Garner VE, Gonzalez-Munoz A, Clark J, Veijola R, Cubbon R, et al. Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity. Nat Genet. 2007;39:329–337. doi:10.1038/ng1958.
  • Hunter K, Rainbow D, Plagnol V, Todd JA, Peterson LB, Wicker LS. Interactions between Idd5.1/Ctla4 and other type 1 diabetes genes. J Immunol. 2007;179:8341–8349.
  • Kostic AD, Gevers D, Siljander H, Vatanen T, Hyötyläinen T, Hämäläinen AM, Peet A, Tillmann V, Pöhö P, Mattila I, et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe. 2015;17:260–273. doi:10.1016/j.chom.2015.01.001.
  • Alkanani AK, Hara N, Gottlieb PA, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. Alterations in intestinal microbiota correlate with susceptibility to type 1 diabetes. Diabetes. 2015;64:3510–3520. doi:10.2337/db14-1847.
  • Endesfelder D, Zu Castell W, Ardissone A, Davis-Richardson AG, Achenbach P, Hagen M, Pflueger M, Gano KA, Fagen JR, Drew JC, et al. Compromised gut microbiota networks in children with anti-islet cell autoimmunity. Diabetes. 2014;63:2006–2014. doi:10.2337/db13-1676.
  • Pinto E, Anselmo M, Calha M, Bottrill A, Duarte I, Andrew PW, Faleiro ML. The intestinal proteome of diabetic and control children is enriched with different microbial and host proteins. Microbiology. 2017;163:161–174. doi:10.1099/mic.0.000412.
  • Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014;5:461. doi:10.3389/fimmu.2014.00461.
  • Lien E, Means TK, Heine H, Yoshimura A, Kusumoto S, Fukase K, Fenton MJ, Oikawa M, Qureshi N, Monks B, et al. Toll-like receptor 4 imparts ligand-specific recognition of bacterial lipopolysaccharide. J Clin Invest. 2000;105:497–504. doi:10.1172/JCI8541.
  • Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801. doi:10.1016/j.cell.2006.02.015.
  • Hoebe K, Janssen EM, Kim SO, Alexopoulou L, Flavell RA, Han J, Beutler B. Upregulation of costimulatory molecules induced by lipopolysaccharide and double-stranded RNA occurs by Trif-dependent and Trif-independent pathways. Nat Immunol. 2003;4:1223–1229. doi:10.1038/ni1010.
  • Hoebe K, Du X, Georgel P, Janssen E, Tabeta K, Kim SO, Goode J, Lin P, Mann N, Mudd S, et al. Identification of Lps2 as a key transducer of MyD88-independent TIR signalling. Nature. 2003;424:743–748. doi:10.1038/nature01889.
  • Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, Takeuchi O, Sugiyama M, Okabe M, Takeda K, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science. 2003;301:640–643. doi:10.1126/science.1087262.
  • Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2:675–680. doi:10.1038/90609.
  • Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, Eng JK, Akira S, Underhill DM, Aderem A. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001;410:1099–1103. doi:10.1038/35074106.
  • Hajjar AM, Ernst RK, Tsai JH, Wilson CB, Miller SI. Human Toll-like receptor 4 recognizes host-specific LPS modifications. Nat Immunol. 2002;3:354–359. doi:10.1038/ni777.
  • Means TK, Hayashi F, Smith KD, Aderem A, Luster AD. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol. 2003;170:5165–5175.
  • Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 2008;455:1109–1113. doi:10.1038/nature07336.
  • Burrows MP, Volchkov P, Kobayashi KS, Chervonsky AV. Microbiota regulates type 1 diabetes through Toll-like receptors. Proc Natl Acad Sci U S A. 2015;112:9973–9977. doi:10.1073/pnas.1508740112.
  • Kondrashova A, Seiskari T, Ilonen J, Knip M, Hyöty H. The ‘Hygiene hypothesis’ and the sharp gradient in the incidence of autoimmune and allergic diseases between Russian Karelia and Finland. APMIS. 2013;121:478–493. doi:10.1111/apm.12023.
  • Kondrashova A, Mustalahti K, Kaukinen K, Viskari H, Volodicheva V, Haapala AM, Ilonen J, Knip M, Mäki M, Hyöty H. Lower economic status and inferior hygienic environment may protect against celiac disease. Ann Med. 2008;40:223–231. doi:10.1080/07853890701678689.
  • Kondrashova A, Viskari H, Haapala AM, Seiskari T, Kulmala P, Ilonen J, Knip M, Hyöty H. Serological evidence of thyroid autoimmunity among schoolchildren in two different socioeconomic environments. J Clin Endocrinol Metab. 2008;93:729–734. doi:10.1210/jc.2007-1644.
  • Richer MJ, Lavallée DJ, Shanina I, Horwitz MS. Toll-like receptor 3 signaling on macrophages is required for survival following coxsackievirus B4 infection. PLoS One. 2009;4:e4127. doi:10.1371/journal.pone.0004127.
  • McCall KD, Thuma JR, Courreges MC, Benencia F, James CB, Malgor R, Kantake N, Mudd W, Denlinger N, Nolan B, et al. Toll-like receptor 3 is critical for coxsackievirus B4-induced type 1 diabetes in female NOD mice. Endocrinology. 2015;156:453–461. doi:10.1210/en.2013-2006.
  • Lang KS, Recher M, Junt T, Navarini AA, Harris NL, Freigang S, Odermatt B, Conrad C, Ittner LM, Bauer S, et al. Toll-like receptor engagement converts T-cell autoreactivity into overt autoimmune disease. Nat Med. 2005;11:138–145. doi:10.1038/nm1176.
  • Zipris D, Lien E, Xie JX, Greiner DL, Mordes JP, Rossini AA. TLR activation synergizes with Kilham rat virus infection to induce diabetes in BBDR rats. J Immunol. 2005;174:131–142.
  • Alkanani AK, Hara N, Lien E, Ir D, Kotter CV, Robertson CE, Wagner BD, Frank DN, Zipris D. Induction of diabetes in the RIP-B7.1 mouse model is critically dependent on TLR3 and MyD88 pathways and is associated with alterations in the intestinal microbiome. Diabetes. 2014;63:619–631. doi:10.2337/db13-1007.
  • Tai N, Wong FS, Wen L. TLR9 deficiency promotes CD73 expression in T cells and diabetes protection in nonobese diabetic mice. J Immunol. 2013;191:2926–2937. doi:10.4049/jimmunol.1300547.
  • Gülden E, Chao C, Tai N, Pearson JA, Peng J, Majewska-Szczepanik M, Zhou Z, Wong FS, Wen L. TRIF deficiency protects non-obese diabetic mice from type 1 diabetes by modulating the gut microbiota and dendritic cells. J Autoimmun. 2018. doi:10.1016/j.jaut.2018.06.003.
  • Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411:603–606. doi:10.1038/35079114.
  • Hugot JP, Chamaillard M, Zouali H, Lesage S, Cézard JP, Belaiche J, Almer S, Tysk C, O’Morain CA, Gassull M, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411:599–603. doi:10.1038/35079107.
  • Li YY, Pearson JA, Chao C, Peng J, Zhang X, Zhou Z, Liu Y, Wong FS, Wen L. Nucleotide-binding oligomerization domain-containing protein 2 (Nod2) modulates T1DM susceptibility by gut microbiota. J Autoimmun. 2017;82:85–95. doi:10.1016/j.jaut.2017.05.007.
  • Hu C, Ding H, Li Y, Pearson JA, Zhang X, Flavell RA, Wong FS, Wen L. NLRP3 deficiency protects from type 1 diabetes through the regulation of chemotaxis into the pancreatic islets. Proc Natl Acad Sci U S A. 2015;112:11318–11323. doi:10.1073/pnas.1513509112.
  • Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt W-D, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009;31:502–512. doi:10.1016/j.immuni.2009.06.025.
  • Denning TL, Norris BA, Medina-Contreras O, Manicassamy S, Geem D, Madan R, Karp CL, Pulendran B. Functional specializations of intestinal dendritic cell and macrophage subsets that control Th17 and regulatory T cell responses are dependent on the T cell/APC ratio, source of mouse strain, and regional localization. J Immunol. 2011;187:733–747. doi:10.4049/jimmunol.1002701.
  • Lelouard H, Fallet M, De Bovis B, Méresse S, Gorvel JP. Peyer’s patch dendritic cells sample antigens by extending dendrites through M cell-specific transcellular pores. Gastroenterology. 2012;142:592–601.e3. doi:10.1053/j.gastro.2011.11.039.
  • Farache J, Koren I, Milo I, Gurevich I, Kim KW, Zigmond E, Furtado GC, Lira SA, Shakhar G. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity. 2013;38:581–595. doi:10.1016/j.immuni.2013.01.009.
  • McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012;483:345–349. doi:10.1038/nature10863.
  • Cerovic V, Houston SA, Scott CL, Aumeunier A, Yrlid U, Mowat AM, Milling SWF. Intestinal CD103(-) dendritic cells migrate in lymph and prime effector T cells. Mucosal Immunol. 2013;6:104–113. doi:10.1038/mi.2012.53.
  • Krutzik SR, Tan B, Li H, Ochoa MT, Liu PT, Sharfstein SE, Graeber TG, Sieling PA, Liu Y-J, Rea TH, et al. TLR activation triggers the rapid differentiation of monocytes into macrophages and dendritic cells. Nat Med. 2005;11:653–660. doi:10.1038/nm1246.
  • Chieppa M, Rescigno M, Huang AY, Germain RN. Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement. J Exp Med. 2006;203:2841–2852. doi:10.1084/jem.20061884.
  • Shen H, Tesar BM, Walker WE, Goldstein DR. Dual signaling of MyD88 and TRIF is critical for maximal TLR4-induced dendritic cell maturation. J Immunol. 2008;181:1849–1858.
  • Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, Nishiyama M, Sato S, Tsujimura T, Yamamoto M, Yokota Y, et al. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat Immunol. 2008;9:769–776. doi:10.1038/ni.1622.
  • Fujimoto K, Karuppuchamy T, Takemura N, Shimohigoshi M, Machida T, Haseda Y, Aoshi T, Ishii KJ, Akira S, Uematsu S. A new subset of CD103+CD8alpha+ dendritic cells in the small intestine expresses TLR3, TLR7, and TLR9 and induces Th1 response and CTL activity. J Immunol. 2011;186:6287–6295. doi:10.4049/jimmunol.1004036.
  • Hu W, Jain A, Gao Y, Dozmorov IM, Mandraju R, Wakeland EK, Pasare C. Differential outcome of TRIF-mediated signaling in TLR4 and TLR3 induced DC maturation. Proc Natl Acad Sci U S A. 2015;112(45):13994–13999.
  • Liu H, Chen F, Wu W, Cao AT, Xue X, Yao S, Evans-Marin HL, Li Y-Q, Cong Y. TLR5 mediates CD172α(+) intestinal lamina propria dendritic cell induction of Th17 cells. Sci Rep. 2016;6:22040. doi:10.1038/srep22040.
  • Mbongue J, Nicholas D, Firek A, Langridge W. The role of dendritic cells in tissue-specific autoimmunity. J Immunol Res. 2014;2014:857143. doi:10.1155/2014/394127.
  • Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, Lehuen A. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med. 2013;19:65–73. doi:10.1038/nm.3042.
  • Xia CQ, Peng R, Chernatynskaya AV, Yuan L, Carter C, Valentine J, Sobel E, Atkinson MA, Clare-Salzler MJ. Increased IFN-α-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-α production. J Immunol. 2014;193:1024–1034. doi:10.4049/jimmunol.1303230.
  • Turley SJ, Lee JW, Dutton-Swain N, Mathis D, Benoist C. Endocrine self and gut non-self intersect in the pancreatic lymph nodes. Proc Natl Acad Sci U S A. 2005;102:17729–17733. doi:10.1073/pnas.0509006102.
  • Tai N, Peng J, Liu F, Gulden E, Hu Y, Zhang X, Chen L, Wong FS, Wen L. Microbial antigen mimics activate diabetogenic CD8 T cells in NOD mice. J Exp Med. 2016;213:2129–2146. doi:10.1084/jem.20160526.
  • Harada Y, Muramatsu M, Shibata T, Honjo T, Kuroda K. Unmutated immunoglobulin M can protect mice from death by influenza virus infection. J Exp Med. 2003;197:1779–1785. doi:10.1084/jem.20021457.
  • Jayasekera JP, Moseman EA, Carroll MC. Natural antibody and complement mediate neutralization of influenza virus in the absence of prior immunity. J Virol. 2007;81:3487–3494. doi:10.1128/JVI.02128-06.
  • Jones DD, DeIulio GA, Winslow GM. Antigen-driven induction of polyreactive IgM during intracellular bacterial infection. J Immunol. 2012;189:1440–1447. doi:10.4049/jimmunol.1200878.
  • Brandtzaeg P, Johansen FE. Mucosal B cells: phenotypic characteristics, transcriptional regulation, and homing properties. Immunol Rev. 2005;206:32–63. doi:10.1111/j.0105-2896.2005.00283.x.
  • Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science. 2004;303:1662–1665. doi:10.1126/science.1091334.
  • Suzuki K, Maruya M, Kawamoto S, Sitnik K, Kitamura H, Agace WW, Fagarasan S. The sensing of environmental stimuli by follicular dendritic cells promotes immunoglobulin A generation in the gut. Immunity. 2010;33:71–83. doi:10.1016/j.immuni.2010.07.003.
  • He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, et al. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity. 2007;26:812–826. doi:10.1016/j.immuni.2007.04.014.
  • Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM. B lymphocytes are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Immunol. 1998;161:3912–3918.
  • Falcone M, Lee J, Patstone G, Yeung B, Sarvetnick N. B lymphocytes are crucial antigen-presenting cells in the pathogenic autoimmune response to GAD65 antigen in nonobese diabetic mice. J Immunol. 1998;161:1163–1168.
  • Mariño E, Tan B, Binge L, Mackay CR, Grey ST. B-cell cross-presentation of autologous antigen precipitates diabetes. Diabetes. 2012;61:2893–2905. doi:10.2337/db12-0006.
  • Palmer JP, Asplin CM, Clemons P, Lyen K, Tatpati O, Raghu PK, Paquette TL. Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science. 1983;222:1337–1339.
  • Baekkeskov S, Aanstoot HJ, Christgau S, Reetz A, Solimena M, Cascalho M, Folli F, Richter-Olesen H, De Camilli P, Camilli PD. Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature. 1990;347:151–156. doi:10.1038/347151a0.
  • Bonifacio E, Lampasona V, Genovese S, Ferrari M, Bosi E. Identification of protein tyrosine phosphatase-like IA2 (islet cell antigen 512) as the insulin-dependent diabetes-related 37/40K autoantigen and a target of islet-cell antibodies. J Immunol. 1995;155:5419–5426.
  • Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb P, Rewers M, Eisenbarth GS, Jensen J, Davidson HW, et al. The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A. 2007;104:17040–17045. doi:10.1073/pnas.0705894104.
  • Rodriguez-Segade S, Camiña MF, Carnero A, Lorenzo MJ, Alban A, Quinteiro C, Lojo S. High serum IgA concentrations in patients with diabetes mellitus: agewise distribution and relation to chronic complications. Clin Chem. 1996;42:1064–1067.
  • Ahmadiafshar A, Mohsenifard MR, Mazloomzadeh S. Evaluation of serum & salivary IgA in patients with type 1 diabetes. PLoS One. 2015;10:e0122757. doi:10.1371/journal.pone.0122757.
  • Conley ME, Delacroix DL. Intravascular and mucosal immunoglobulin A: two separate but related systems of immune defense? Ann Intern Med. 1987;106:892–899.
  • D’Auria G, Peris-Bondia F, Džunková M, Mira A, Collado MC, Latorre A, Moya A. Active and secreted IgA-coated bacterial fractions from the human gut reveal an under-represented microbiota core. Sci Rep. 2013;3:3515. doi:10.1038/srep03515.
  • Palm NW, De Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, Degnan PH, Hu J, Peter I, Zhang W, et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell. 2014;158:1000–1010. doi:10.1016/j.cell.2014.08.006.
  • Zeng MY, Cisalpino D, Varadarajan S, Hellman J, Warren HS, Cascalho M, Inohara N, Núñez G. Gut Microbiota-Induced Immunoglobulin G Controls Systemic Infection by Symbiotic Bacteria and Pathogens. Immunity. 2016;44:647–658. doi:10.1016/j.immuni.2016.02.006.
  • Wilson CS, Elizer SK, Marshall AF, Stocks BT, Moore DJ. Regulation of B lymphocyte responses to Toll-like receptor ligand binding during diabetes prevention in non-obese diabetic (NOD) mice. J Diabetes. 2016;8:120–131. doi:10.1111/1753-0407.12263.
  • Montandon R, Korniotis S, Layseca-Espinosa E, Gras C, Mégret J, Ezine S, Dy M, Zavala F. Innate pro-B-cell progenitors protect against type 1 diabetes by regulating autoimmune effector T cells. Proc Natl Acad Sci U S A. 2013;110:E2199–208. doi:10.1073/pnas.1222446110.
  • Gianchecchi E, Crinò A, Giorda E, Luciano R, Perri V, Russo AL, Cappa M, Rosado MM, Fierabracci A, Pietropaolo M. Altered B cell homeostasis and toll-like receptor 9-driven response in type 1 diabetes carriers of the C1858T PTPN22 allelic variant: implications in the disease pathogenesis. PLoS One. 2014;9:e110755. doi:10.1371/journal.pone.0110755.
  • Wooldridge L, Ekeruche-Makinde J, Van Den Berg HA, Skowera A, Miles JJ, Tan MP, Dolton G, Clement M, Llewellyn-Lacey S, Price DA, et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem. 2012;287:1168–1177. doi:10.1074/jbc.M111.289488.
  • Bowman LM, Holt PG. Selective enhancement of systemic Th1 immunity in immunologically immature rats with an orally administered bacterial extract. Infect Immun. 2001;69:3719–3727. doi:10.1128/IAI.69.6.3719-3727.2001.
  • Feng T, Wang L, Schoeb TR, Elson CO, Cong Y. Microbiota innate stimulation is a prerequisite for T cell spontaneous proliferation and induction of experimental colitis. J Exp Med. 2010;207:1321–1332. doi:10.1084/jem.20092253.
  • Penders J, Thijs C, Van Den Brandt PA, Kummeling I, Snijders B, Stelma F, Adams H, Van Ree R, Stobberingh EE. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut. 2007;56:661–667. doi:10.1136/gut.2006.100164.
  • Huang YJ, Nelson CE, Brodie EL, Desantis TZ, Baek MS, Liu J, Woyke T, Allgaier M, Bristow J, Wiener-Kronish JP, et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J Allergy Clin Immunol. 2011;127:372–81.e1-3. doi:10.1016/j.jaci.2010.10.048.
  • Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–498. doi:10.1016/j.cell.2009.09.033.
  • Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4615–4622. doi:10.1073/pnas.1000082107.
  • Teng F, Klinger CN, Felix KM, Bradley CP, Wu E, Tran NL, Umesaki Y, Wu H-JJ. Gut Microbiota Drive Autoimmune Arthritis by Promoting Differentiation and Migration of Peyer’s Patch T Follicular Helper Cells. Immunity. 2016;44:875–888. doi:10.1016/j.immuni.2016.03.013.
  • Nakae S, Nambu A, Sudo K, Iwakura Y. Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. J Immunol. 2003;171:6173–6177.
  • Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–240. doi:10.1084/jem.20041257.
  • Kriegel MA, Sefik E, Hill JA, Wu HJ, Benoist C, Mathis D. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci U S A. 2011;108:11548–11553. doi:10.1073/pnas.1108924108.
  • Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky AV. Gender bias in autoimmunity is influenced by microbiota. Immunity. 2013;39:400–412. doi:10.1016/j.immuni.2013.08.013.
  • Kenefeck R, Wang CJ, Kapadi T, Wardzinski L, Attridge K, Clough LE, Heuts F, Kogimtzis A, Patel S, Rosenthal M, et al. Follicular helper T cell signature in type 1 diabetes. J Clin Invest. 2015;125:292–303. doi:10.1172/JCI76238.
  • Ferreira RC, Simons HZ, Thompson WS, Cutler AJ, Dopico XC, Smyth DJ, Mashar M, Schuilenburg H, Walker NM, Dunger DB, et al. IL-21 production by CD4+ effector T cells and frequency of circulating follicular helper T cells are increased in type 1 diabetes patients. Diabetologia. 2015;58:781–790. doi:10.1007/s00125-015-3509-8.
  • Wan X, Thomas JW, Unanue ER. Class-switched anti-insulin antibodies originate from unconventional antigen presentation in multiple lymphoid sites. J Exp Med. 2016;213:967–978. doi:10.1084/jem.20151869.
  • Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–1061. doi:10.1126/science.1079490.
  • Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol. 2010;11:7–13. doi:10.1038/ni.1818.
  • Gershon RK, Kondo K. Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology. 1970;18:723–737.
  • Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73. doi:10.1038/83784.
  • Godfrey VL, Wilkinson JE, Rinchik EM, Russell LB. Fatal lymphoreticular disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic environment: potential model for thymic education. Proc Natl Acad Sci U S A. 1991;88:5528–5532.
  • Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. Am J Pathol. 1991;138:1379–1387.
  • Caramalho I, Lopes-Carvalho T, Ostler D, Zelenay S, Haury M, Demengeot J. Regulatory T cells selectively express toll-like receptors and are activated by lipopolysaccharide. J Exp Med. 2003;197:403–411.
  • Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341. doi:10.1126/science.1198469.
  • Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232–236. doi:10.1038/nature12331.
  • Geuking MB, Cahenzli J, Lawson MA, Ng DC, Slack E, Hapfelmeier S, McCoy KD, Macpherson AJ. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 2011;34:794–806. doi:10.1016/j.immuni.2011.03.021.
  • Rahman MK, Midtling EH, Svingen PA, Xiong Y, Bell MP, Tung J, Smyrk T, Egan LJ, Faubion WA. The pathogen recognition receptor NOD2 regulates human FOXP3+ T cell survival. J Immunol. 2010;184:7247–7256. doi:10.4049/jimmunol.0901479.
  • Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, Von Bergen M, McCoy KD, Macpherson AJ, Danska JS. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339:1084–1088. doi:10.1126/science.1233521.
  • Roediger WE. Utilization of nutrients by isolated epithelial cells of the rat colon. Gastroenterology. 1982;83:424–429.
  • Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, Bultman SJ. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–526. doi:10.1016/j.cmet.2011.02.018.
  • Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr. 2009;139:1619–1625. doi:10.3945/jn.109.104638.
  • Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe. 2015;17:662–671. doi:10.1016/j.chom.2015.03.005.
  • Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–573. doi:10.1126/science.1241165.
  • Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–455. doi:10.1038/nature12726.
  • Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–450. doi:10.1038/nature12721.
  • Croset M, Rajas F, Zitoun C, Hurot JM, Montano S, Mithieux G. Rat small intestine is an insulin-sensitive gluconeogenic organ. Diabetes. 2001;50:740–746.
  • de Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Bäckhed F, Mithieux G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156:84–96. doi:10.1016/j.cell.2013.12.016.
  • Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286. doi:10.1038/nature08530.
  • Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci U S A. 2008;105:16767–16772. doi:10.1073/pnas.0808567105.
  • Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, Hardt PD. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 2010;18:190–195. doi:10.1038/oby.2009.167.
  • Mariño E, Richards JL, McLeod KH, Stanley D, Yap YA, Knight J, McKenzie C, Kranich J, Oliveira AC, Rossello FJ, et al. Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat Immunol. 2017;18:552–562. doi:10.1038/ni.3713.
  • Sun J, Furio L, Mecheri R, van der Does AM, Lundeberg E, Saveanu L, Chen Y, van Endert P, Agerberth B, Diana J. Pancreatic β-cells Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial peptide Expressed under the Influence of the Gut Microbiota. Immunity. 2015;43(2):304–317.
  • Livanos AE, Greiner TU, Vangay P, Pathmasiri W, Stewart D, McRitchie S, Li H, Chung J, Sohn J, Kim S, et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat Microbiol. 2016;1:16140. doi:10.1038/nmicrobiol.2016.140.
  • Brown K, Godovannyi A, Ma C, Zhang Y, Ahmadi-Vand Z, Dai C, Gorzelak MA, Chan Y, Chan JM, Lochner A, et al. Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice. ISME J. 2016;10:321–332. doi:10.1038/ismej.2015.114.
  • de Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–14696. doi:10.1073/pnas.1005963107.
  • Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529:212–215. doi:10.1038/nature16504.
  • Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–108. doi:10.1126/science.1208344.
  • Walker AW, Duncan SH, McWilliam Leitch EC, Child MW, Flint HJ. pH and peptide supply can radically alter bacterial populations and short-chain fatty acid ratios within microbial communities from the human colon. Appl Environ Microbiol. 2005;71:3692–3700. doi:10.1128/AEM.71.7.3692-3700.2005.
  • Kovatcheva-Datchary P, Nilsson A, Akrami R, Lee YS, de Vadder F, Arora T, Hallen A, Martens E, Björck I, Bäckhed F. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of prevotella. Cell Metab. 2015;22:971–982. doi:10.1016/j.cmet.2015.10.001.
  • Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214. doi:10.1126/science.1241214.
  • Reddy S, Bibby NJ, Elliott RB. Early nicotinamide treatment in the NOD mouse: effects on diabetes and insulitis suppression and autoantibody levels. Diabetes Res. 1990;15:95–102.
  • Reddy S, Bibby NJ, Wu D, Swinney C, Barrow G, Elliott RB. A combined casein-free-nicotinamide diet prevents diabetes in the NOD mouse with minimum insulitis. Diabetes Res Clin Pract. 1995;29:83–92.
  • Mathieu C, Laureys J, Sobis H, Vandeputte M, Waer M, Bouillon R. 1,25-Dihydroxyvitamin D3 prevents insulitis in NOD mice. Diabetes. 1992;41:1491–1495.
  • Mathieu C, Waer M, Laureys J, Rutgeerts O, Bouillon R. Prevention of autoimmune diabetes in NOD mice by 1,25 dihydroxyvitamin D3. Diabetologia. 1994;37:552–558.
  • Takiishi T, Ding L, Baeke F, Spagnuolo I, Sebastiani G, Laureys J, Verstuyf A, Carmeliet G, Dotta F, Van Belle TL, et al. Dietary supplementation with high doses of regular vitamin D3 safely reduces diabetes incidence in NOD mice when given early and long term. Diabetes. 2014;63:2026–2036. doi:10.2337/db13-1559.
  • Rewers M, Liu E, Simmons J, Redondo MJ, Hoffenberg EJ. Celiac disease associated with type 1 diabetes mellitus. Endocrinol Metab Clin North Am. 2004;33:197–214, xi. doi:10.1016/j.ecl.2003.12.007.
  • Rewers M, Eisenbarth GS. Autoimmunity: celiac disease in T1DM-the need to look long term. Nat Rev Endocrinol. 2011;8:7–8. doi:10.1038/nrendo.2011.193.
  • Brorsson CA, Pociot F, Consortium TDG. Shared genetic basis for type 1 diabetes, islet autoantibodies, and autoantibodies associated with other immune-mediated diseases in families with type 1 diabetes. Diabetes Care. 2015;38(Suppl 2):S8–13. doi:10.2337/dcs15-2003.
  • Maglio M, Florian F, Vecchiet M, Auricchio R, Paparo F, Spadaro R, Zanzi D, Rapacciuolo L, Franzese A, Sblattero D, et al. Majority of children with type 1 diabetes produce and deposit anti-tissue transglutaminase antibodies in the small intestine. Diabetes. 2009;58:1578–1584. doi:10.2337/db08-0962.
  • Hansen AK, Ling F, Kaas A, Funda DP, Farlov H, Buschard K. Diabetes preventive gluten-free diet decreases the number of caecal bacteria in non-obese diabetic mice. Diabetes Metab Res Rev. 2006;22:220–225. doi:10.1002/dmrr.609.
  • Marietta EV, Gomez AM, Yeoman C, Tilahun AY, Clark CR, Luckey DH, Murray JA, White BA, Kudva YC, Rajagopalan G, et al. Low incidence of spontaneous type 1 diabetes in non-obese diabetic mice raised on gluten-free diets is associated with changes in the intestinal microbiome. PLoS One. 2013;8:e78687. doi:10.1371/journal.pone.0078687.
  • Funda DP, Kaas A, Tlaskalová-Hogenová H, Buschard K. Gluten-free but also gluten-enriched (gluten+) diet prevent diabetes in NOD mice; the gluten enigma in type 1 diabetes. Diabetes Metab Res Rev. 2008;24:59–63. doi:10.1002/dmrr.748.
  • Hummel S, Pflüger M, Hummel M, Bonifacio E, Ziegler AG. Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study. Diabetes Care. 2011;34:1301–1305. doi:10.2337/dc10-2456.
  • Hu Y, Jin P, Peng J, Zhang X, Wong FS, Wen L. Different immunological responses to early-life antibiotic exposure affecting autoimmune diabetes development in NOD mice. J Autoimmun. 2016;72:47–56. doi:10.1016/j.jaut.2016.05.001.
  • Candon S, Perez-Arroyo A, Marquet C, Valette F, Foray AP, Pelletier B, Milani C, Ventura M, Bach J-F, Chatenoud L, et al. Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes. PLoS One. 2015;10:e0125448. doi:10.1371/journal.pone.0125448.
  • Hansen CH, Krych L, Nielsen DS, Vogensen FK, Hansen LH, Sørensen SJ, Buschard K, Hansen AK. Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse. Diabetologia. 2012;55:2285–2294. doi:10.1007/s00125-012-2564-7.
  • Tormo-Badia N, Håkansson A, Vasudevan K, Molin G, Ahrné S, Cilio CM. Antibiotic treatment of pregnant non-obese diabetic (NOD) mice leads to altered gut microbiota and intestinal immunological changes in the offspring. Scand J Immunol. 2014;80:250–260. doi:10.1111/sji.12205.
  • Hu Y, Peng J, Tai N, Hu C, Zhang X, Wong FS, Wen L. Maternal antibiotic treatment protects offspring from diabetes development in nonobese diabetic mice by generation of tolerogenic APCs. J Immunol. 2015;195:4176–4184. doi:10.4049/jimmunol.1500884.
  • Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, Baccaglini L, Mohapatra A, Mohapatra SS, Misra PR, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548:407–412. doi:10.1038/nature23480.
  • Sanders ME. Impact of probiotics on colonizing microbiota of the gut. J Clin Gastroenterol. 2011;45(Suppl):S115–9. doi:10.1097/MCG.0b013e318227414a.
  • Matsuzaki T, Nagata Y, Kado S, Uchida K, Kato I, Hashimoto S, Yokokura T. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of Lactobacillus casei. APMIS. 1997;105:643–649.
  • Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, Marzotti S, Corneli RB, Ferretti E, Gulino A, Grasso F, et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia. 2005;48:1565–1575. doi:10.1007/s00125-005-1831-2.
  • Valladares R, Sankar D, Li N, Williams E, Lai KK, Abdelgeliel AS, Gonzalez CF, Wasserfall CH, Larkin J, Schatz D, et al. Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS One. 2010;5:e10507. doi:10.1371/journal.pone.0010507.
  • Dolpady J, Sorini C, Di Pietro C, Cosorich I, Ferrarese R, Saita D, Clementi M, Canducci F, Falcone M. Oral probiotic VSL#3 prevents autoimmune diabetes by modulating microbiota and promoting indoleamine 2,3-dioxygenase-enriched tolerogenic intestinal environment. J Diabetes Res. 2016;2016:7569431. doi:10.1155/2016/7569431.
  • Hänninen A, Toivonen R, Pöysti S, Belzer C, Plovier H, Ouwerkerk JP, Emani R, Cani PD, De Vos WM. induces gut microbiota remodelling and controls islet autoimmunity in NOD mice. Gut. 2018;67:1445–1453. doi:10.1136/gutjnl-2017-314508.
  • Jia L, Shan K, Pan LL, Feng N, Lv Z, Sun Y, Li J, Wu C, Zhang H, Chen W, et al. CGMCC0313.1 protects against autoimmune diabetes by modulating intestinal immune homeostasis and inducing pancreatic regulatory T cells. Front Immunol. 2017;8:1345. doi:10.3389/fimmu.2017.01345.
  • Primec M, Klemenak M, Di Gioia D, Aloisio I, Bozzi Cionci N, Quagliariello A, Gorenjak M, Mičetić-Turk D, Langerholc T. Clinical intervention using Bifidobacterium strains in celiac disease children reveals novel microbial modulators of TNF-α and short-chain fatty acids. Clin Nutr. 2018;S0261-5614(18):31150–31160.
  • Egshatyan L, Kashtanova D, Popenko A, Tkacheva O, Tyakht A, Alexeev D, Karamnova N, Kostryukova E, Babenko V, Vakhitova M, et al. Gut microbiota and diet in patients with different glucose tolerance. Endocr Connect. 2016;5:1–9. doi:10.1530/EC-15-0094.
  • Marcial GE, Ford AL, Haller MJ, Gezan SA, Harrison NA, Cai D, Meyer JL, Perry DJ, Atkinson MA, Wasserfall CH, et al. Lactobacillus johnsonii N6.2 modulates the host immune responses: a double-blind, randomized trial in healthy adults. Front Immunol. 2017;8:655. doi:10.3389/fimmu.2017.00655.
  • Salami F, Abels M, Hyöty H, Vaziri-Sani F, Aronsson C, Vehik K, Delli A, Hagopian W, Rewers M, Ziegler A, et al. Detection of Lactobacilli in monthly mail-in stool samples from 3-18 months old infant at genetic risk for Type 1 Diabetes. Int J Probiotics Prebiotics. 2012;7:135–144.
  • Bowden TA, Mansberger AR, Lykins LE. Pseudomembraneous enterocolitis: mechanism for restoring floral homeostasis. Am Surg. 1981;47:178–183.
  • Shi Y, Dong Y, Huang W, Zhu D, Mao H, Su P. Fecal microbiota transplantation for ulcerative colitis: a systematic review and meta-analysis. PLoS One. 2016;11:e0157259. doi:10.1371/journal.pone.0157259.
  • Kootte RS, Levin E, Salojärvi J, Smits LP, Hartstra AV, Udayappan SD, Hermes G, Bouter KE, Koopen AM, Holst JJ, et al. Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metab. 2017;26:611–9.e6. doi:10.1016/j.cmet.2017.09.008.
  • Peng J, Narasimhan S, Marchesi JR, Benson A, Wong FS, Wen L. Long term effect of gut microbiota transfer on diabetes development. J Autoimmun. 2014;53:85–94. doi:10.1016/j.jaut.2014.03.005.
  • Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273. doi:10.1126/science.1223490.
  • Wong FS, Hu C, Zhang L, Du W, Alexopoulou L, Flavell RA, Wen L. The role of Toll-like receptors 3 and 9 in the development of autoimmune diabetes in NOD mice. Ann N Y Acad Sci. 2008;1150:146–148. doi:10.1196/annals.1447.039.
  • Hebbandi Nanjundappa R, Ronchi F, Wang J, Clemente-Casares X, Yamanouchi J, Sokke Umeshappa C, Yang Y, Blanco J, Bassolas-Molina H, Salas A, et al. A gut microbial mimic that hijacks diabetogenic autoreactivity to suppress colitis. Cell. 2017;171:655–67.e17. doi:10.1016/j.cell.2017.09.022.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.