Are we Any Close to Unraveling the Mechanism of Interactions Among Susceptibility Genes Towards Type 1 Diabetes, Gut Microbiota Along with Environmental Factors , Specifically Early Diet Patterns –A S

Authors

Kulvinder Kochar Kaur 1*, Gautam Allahbadia 2, Mandeep Singh 3
1 Scientific Director, Kulvinder  Kaur Centre For Human Reproduction, 721,G.T.B. Nagar, Jalandhar-144001, Punjab, India
2Scientific Director, Ex-Rotunda-A Centre for Human Reproduction, 672,Kalpak Garden,Perry Cross Road, Near Otter’s Club,Bandra(W)-400040 Mumbai, India
3Consultant Neurologist, Swami Satyanand Hospital, Near Nawi Kachehri,Baradri,, Ladowali road,JALANDHAR, Punjab India.
*Corresponding author: Kulvinder Kochar Kaur, Scientific Director, Kulvinder  Kaur Centre For Human Reproduction, 721,G.T.B. Nagar, Jalandhar-144001, Punjab, India

Article Information

*Corresponding author: Kulvinder Kochar Kaur, Scientific Director, Kulvinder  Kaur Centre For Human Reproduction, 721,G.T.B. Nagar, Jalandhar-144001, Punjab, India

Accepted date: February 09, 2021
Accepted date: February 18, 2021
published date: February 24, 2021

Citation: Kulvinder K Kaur, Allahbadia G, Singh M. “Are we Any Close to Unraveling the Mechanism of Interactions Among Susceptibility Genes Towards Type 1 Diabetes, Gut Microbiota Along with Environmental Factors , Specifically Early Diet Patterns –A Systematic Review.’’. Endocrinology and Surgical Endocrinology, 2(1); DOI: http;//doi.org/03.2021/1.1005.
Copyright: © 2021 Kulvinder Kochar Kaur. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly Cited.

Abstract

Earlier wehad reviewed on the aetiopathogenesis of   Type 1 diabetes mellitus(T1D) along with role of gutmicrobiota(GM) ,genes ,immunotherapies besides the role ofGM in obesity ,type 2 diabetes and probiotics in detail. Whereas the pathogens for autoimmune diseases continue to be mostly not clear ,genetic proneness  as  well as environmental factors   have been believed  to be the main etiologies.Of the    environmental factors  the, microbiome is associated  with  autoimmune diseases through direct  as  well as      indirect  crosstalk with innate  as  well as      adaptive  immune cells .This leads to loss of   immune tolerance ,chronic inflammation   as  well as    immune response     against  host tissues .The posited part of  microbiome  in  autoimmunity are Molecular mimicry ,epitope spreading ,bystander activation, as  well as      continued infection.Further the longitudinal studies have pointed toward the implication of geographical variations .Here we decided to conduct a systematic review  on the role of gut microbiota and its relation with Type 1 diabetes mellitus,interaction with other environmental factors like  delivery mode ,mode of baby feeding and its impact on  GMlike use of breast feeding only at least till 4 mths  ,Ultimately it has been observed that delaying gluten introduction till 4mths as  well as     cows milk beyond 12mths of age along with addition of early pre/probiotics  in those children possessing high risk susceptibility genes .More work is required to evaluate gut virome and other components like archeome ,Microbiota of vagina ,skin  as  well as     metabolome to arrive at a conclusion .Moreover use of diets like Mediterranean diet ,FUN2 as  well as       ArH targeting to avoid generation of T1D needs to be exploited.


Keywords: gutmicrobiota(GM); Type 1 diabetes mellitus(T1D);probiotics;vaginal delivery ;breast feeding

1. Introduction

Earlier wehad reviewed on the aetiopathogenesis of   Type 1 diabetes mellitus(T1D) along with role of gutmicrobiota(GM) ,genes ,immunotherapies along with role ofGM in obesity ,type 2 diabetes and probiotics in detail[1-12] .The human microbiome  comprises of trillions of bacterial ,viral as  well as     fungal microorganisms[13].Coevolution took place in a symbiotic fashion along with humans for 100-1000’s of years[14].Why so much variation of these microorganisms occurs is secondary to host lifestyle  ,geographical placement ,dietary habits,infections ,sex,age as  well as     genetic background[15].It has been demonstrated that the human microbiome has the capacity to influence the host physiology in multiple ways like metabolism , immunity , along with behavior[16].Thus interfering with human microbiota  might cause disease.Multiple studies have ,illustrated that the human microbiome   has the capacity  to influence the etiopathogenesis  of  immune diseases, Specifically autoimmune diseases,where the immune system has the inability of differentiating self from the nonself  proteins as  well as  attack  self tissues .Like in Multiple Sclerosis (MS)[16], Rheumatoid arthritis( RA), systemic lupus erythematosus(SLE), antiphospholipid syndrome( APS)[17], Crohn’s disease(CD)   [18], Ulcerative colitis (UC)[19], inflammatory bowel disease (IBD)[20].Coeliac  disease[12], Type 1 diabetes mellitus(T1D) [22,23].

The existence of particular   Gut Microbiota(GM) as  well as      total diversity ,are key in generation of the nascent immune system.Studies carried out in germ free( GF)  as  well as      gnotobiotic  mouse models demonstrated  significant part in the manipulation as well as     differentiation  of innate immune  cell kinds  particularly  interleukin(IL)-17 Generating     CD4+T cells (Th17cells) as  well as      Foxp3 + regulatory T(Treg)cells[24]. Particularly segmentous filamentous bacteria(SFB) are believed to stimulate the expression of pro inflammatory  Th17cells, significant   for sustaining the mucosal barrier as  well as     , confer protection  to non obese diabetic(NOD)  mice from Generating  T1D[24,25]. Nevertheless,in other mouse models  of autoimmune diseases(like K/BxN mouse models  OF  autoimmune arthritis ). SFB are documented to facilitate disease propagation  through significant  Th17 collection ,pointing to their part in autoimmunity is etiologically particular  [26].Other bacteria confer protection   like  Lactobacillus, Bifidobacterium as  well as      Clostridium  species,are responsible for the stimulation of ant- inflammatory  Treg)cells  whereas Bacteroides fragilis polysaccharide A(PSA) stimulates  IL-10  generation as  well as      is understood to repress  Th17cells  responses[27].

Whereas the pathogens for autoimmune diseases continue to be mostly not clear ,genetic proneness [28] as  well as  environmental factors  [29] have been believed  to be the main etiologies.Of the    environmental factors  the, microbiome is associated  with  autoimmune diseases through direct  as  well as      indirect  crosstalk with innate  as  well as      adaptive  immune cells .This leads to loss of   immune tolerance ,chronic inflammation   as  well as    immune response     against  host tissues [30].The posited part of  microbiome  in  autoimmunity are Molecular mimicry ,epitope spreading ,bystander activation, as  well as      continued infection[30,31].Hence the immune tolerance loss  might get stimulated  changes in  microbiome  composition .Various studies  have actually illustrated that the gut   microbiome composition of patients with    autoimmune diseases is markedly separate in contrast to   healthy subjects[32-35].

Leaving few exceptions (like Sardinia ,Italy ), autoimmune diseases incidence  keeps a north-south wave ,with escalated incidences observed in Nordic countries ,like Finland,Sweden  as  well as       Norway [36,37].GM studies on the influence of extreme climatic situations (like polar expeditions )[38] as  well as      birth month/place [39] reveal how climate, Specifically sunlight exposure ,influence GM composition as  well as      immune impairment.The  factors   that are significant in northern population are circadian rhythm impairment as  well as      Viamin D deficiencies ,that have also been illustrated to lead to immune impairment through a swing in the GM ,resulting in autoimmune diseases like T1D[40,41].

T1D is believed to be a disease having the properties of insulin deficiency secondary to autoimmune  injury  or function loss  of the pancreatic insulin generating beta cells  .It is significant  to observe that autoimmunity is found as a major factor resulting in T1D by most researchers ,certain workers believe that autoimmunity occurs following other factors  like endoplasmic reticulum(ER) stress  resulting in beta cells   deletion[42].By historical facts T1D has been believed  to be escalated blood glucose  amounts(hyperglycemia), as  well as      the existence of 1 or greater antibodies,all of which take place/or are existent prior to beta cells   ablation[43]. Autoimmunity  can get mounted against insulin(IAA), glutamic acid decarboxylase((GAD 65),Insulinoma  associated  autoantigen-2(IA2A), as  well as/or zinc transporter8(ZnT8A) along with  may occur many years before symptoms onset[43].Besides the major antibodies,a new found family of neoepitopes Generated with post translation modulation got isolated[44].Hybrid insulin peptides(HIP’s) represent a very intriguing neo peptides which get generated  with the fusing of an insulin neopeptide along with a liberating granule  peptides in the granules[45].The commonest autoantibody   seen before the disease initiation is against IAA,with the IAA amounts association robustly with  the rate of propagation  towards overt T1D in children [43].This event is usually known as seroconversion ,a significant terminology  utilized all through this review.Here we decided to conduct a systematic review on the role of GM in detail with regards to T1D .

Methods

Thus a systematic review was carried out using the pubmed, Web of Science , Medline, Embase, Cochrane reviews,  and Google Scholar, Search  engine with the MeSH Terms; impaired lipid metabolism; oxidative stress;inflammation; ;Gut Microbiota(GM); Type 1 diabetes (T1D);breast feeding ;mode of delivery;gluten foods introduction;role of omics in studying the etiopathogenesis of T1D from 1990’s till date in 2021.

Results

We found a total of 750 articles, out of which we selected 194 articles for this review.No meta-analysis was done. did a search using the pubmed search engine using the MeSH terms

Whereas T1D remains 1 of the maximum represented chronic ailments in childhood ,approximately 25%of individuals diagnosed with this disease compromise of adults [46].There has been an escalation of the incidence worldwide for the past few decades particularly post World war II in the west [47-49].Although great genetic impact  is there the enhancement of T1D prevalence as  well as      varying incidence  rates across various countries , as  well as      even among countries that are just adjacent to each other  in Europe(Russia Karelia  as  well as      Finnish Karelia),points to an interaction among proneness genes  along with particular       environmental  effects[50].Discordant outcomes for T1D twins ,all point that environmental  effects have a great part in the etiopathogenesis of the disease[51].

The non-obese diabetic(NOD) mouse  model is the commonest animal model utilized to study  T1D,in view of considerable akinness to human T1D regards to recognition of autoantigens,immunopathology as  well as      gene proneness[52].Akin to humans in NOD) mouse  model shows T1D correlated variation in MHC Class II that influences the presentation of islet –obtained antigens to the T Cells .Besides that ,a lot of non  MHC  prone genes  that are common both for NOD mice as  well as       humans  are correlated with T1D risk  like protein tyrosine phosphatase non-receptor22(PTPN22)gene, of  cytotoxic lymphocyte associated protein 4 gene(CTLA4), IL2RA(codes-αsubunit of IL2R[53].It was initially seen that colony hygiene influences  the incidence of T1D in NOD mice [54].Other studies on NOD mice have further illustrated that the Gut Microbiome  can crossreact with immune system to control DM pathogenesis in mice [55]. Nevertheless,unlike humans ,female NOD mice  possesses significantly greater   T1D   incidence in contrast to   male NOD mice.A study conducted by Markle etal.,[56] revealed  that male NOD mice   in specific  pathogen  free(SPE) situations were greater protected against     T1D  in contrast to   female NOD mice. Nevertheless,  in germ free   situations,  both male as  well as      female mice had equal incidence  rates.Some   commensal bacteria  that are believed to escalate T   amounts  are also observed to have the capacity  of protecting male NOD mice against  T1D onset [56].Intriguingly ,transfer of GM from male NOD mice to female mice changed  the composition of Gut Microbiome  in recipient mice  and hence protected them from T1D  [56].This  points that changing of Gut Microbiome   can implicate immune system as  well as      pathogenesis of disease .

Inspite of variations in NOD) mouse  model  T1D   etiology  that is apparent,studies conducted in vivo give the mode of action  strategy that is not feasible in human studies having been key in getting insight  in disease onset as well as     propagation.Here longitudinal studies as  well as       those on NOD mice along with role of Gut Microbiome   as it associates to environmental factor in T1D pathogenesis is detailed.

2.Human Longitudinal Studies

Genome –wide association study (GWAS) studies have observed equivalent to 50 genetic areas which implicate the risk of generation of T1D[57].The risk of generation of T1D gets decided by finding the genetic factors   like T1D-correlated SNP in the human leukocyte antigen (HLA )gene ,more particularly ,the HLA-DQ as  well as     HLA-DR  protein coding genes        DQ1 as  well as     DQB1[58]. Nevertheless, just genetic proneness is not enough to reason out the T1D   onset[59].Till now the outcomes from these studies  have been wide as  well as    primarily associative ,pointing to the multiple facets of this disease.

For finding the etiopathogenetic environmental factors   that initiate the disease origin, longitudinal studies of huge at –risk cohorts  are needed  across a broad geographical area.These actions have got started –are Teddy , DABIMMUNE  ,BABYDIET(i.e a substudy  of the huge BABYDIAB),ABIS,TRIGR as  well as     FINDIA(a  substudy in the Type 1 diabetes mellitus Prediction as  well as     Prevention DIPP Study).Here observations of Teddy , DABIMMUNE   as  well as     ABIS,is detailed with concentration   on Gut Microbiome    in the generation of  autoimmunity .

2.1Teddy Study

The environmental determinants of  diabetes in the Young(TEDDY)got designed  for  constantly monitoring the  children  isolated  having a proneness for generation of T1D.The study got carried out in 6 various clinical centers in Colorado ,Washington State,Georgia/Florida  in the US as  well as     Finland ,Germany along with Sweden.From 2004-2010 ,general population(GP) along with first degree relations(FDR)newborns got screened for HLA kinds[58].Regarding all the 6 geographical areas, same high risk haplogenotypes got isolated,of these haplogenotypes were taken into account  for the inclusion particulars for the GP(DR3/4, DR4/4, DR4/8, DR3/3),whereas 9 haplogenotypes were taken into account  for the FDR(DR3/4, DR4/4, DR4/8, DR3/3, DR4/4b, DR4/1, DR4/13, DR4/9 along with DR3/9((https://teddy .epi.usf .edu/research).

2 extra studies got carried out  utilizing TEDDY samples.In the study by Vatanen etal.[23],stool samples  got acquired  as  well as     sequenced from 783 children  mthly at age3mths  till the clinical end stage (seroconversion , autoantibodies  getting found ).In islet autoimmunity(IA),case  control cohorts had a greater prevalence   of Streptococcus  group/mitis/oralis /pneumonia whereas controls possessed greater prevalence   Lactobacillus rhamnosus as  well as     Bifidobacterium    dentium ,2 usually species in probiotic cocktails.In   T1D case  control cohorts  , T1Dcases possessed greater amount of Bifidobacterium    pseudo catenulam,Roseburia hominis , as well as     Alistepes  shahii, whereas healthy controls possessed greater ,Streptococcus thermophilus  as well as     Lactobacillus lactis .Stewart et al.,[60]in another study  evaluated stool samples   from 903 children among 3 as  well as      46mths of age utilizing 16S rRNA  (V4) as well as     carried out  metagenomic sequencing results.A nested case  control Evaluation documented that alpha diversity Microbiata maturation  as  well as        Microbiata by age Z scores(MAZ) were akin among  cases   as  well as        controls  regards to both IA as  well as      T1D cohorts  .The relative excess of maximum escalated genera illustrated  subtle  composition changes  that had a greater prevalence of Erysipelotrichaceae  in IA cases .In contrast to   healthy controls,   T1D cases possessed a greater excess of  Parabacteroides(p<0.001) as  well as      a reduced prevalence of11 genera of Ruminococcacea, Lactococcus, Streptococcus as  well as      Akkermansia.In total TEDDY isolated  various bacteria  weakly associated with      T1D initiation, Nevertheless,future studies  are required  for  finding out the mode of actions working.

2.2 Dabimmune Study

In case of northern Europe , greater variation in  incidence of autoimmune diseases have been demonstrated among adjacent countries .Like despite the frequency of HLA  genotypes  are quite akin among  Finland as  well as      Russian Karelia  ,the   incidence of T1D are 6times greater in Finland[60].These  trends seen are believed  to be based on the national  public healthy standards  along with personal hygiene  methods, as  well as      seen that it is the developed countries  having a greater incidence of autoimmune diseases  like T1D   in contrast to  lesser developed countries.Hygiene as  well as      autoimmunity ( like Hygiene posit ) are correlated with the found reduction in GM diversity  in greater hygienic  surroundings[61-63].The absence of  alpha diversity  is believed to escalate the chances of pathogenic invasion  (like antibiotic stimulated Clostridium difficile  infections  )[64] as  well as      has been demonstrated to accelerate     autoimmunity in persons prone  to same. The DABIMMUNE longitudinal study  tried to isolate  the environmental factors   which could be pointed to have greater chances of autoimmune  as well as     allergenic  diseases.

In 2008,in   DABIMMUNE, recruitment of equivalent to 1000 newborn infants  possessing high risk HLA   haplotypes from Finland,Estonia  as well as      Russia  was done.Blood In  addition to   stool samples as well as     clinical metadata ,got acquired from 1mth to 3yrs of age .The earlier Evaluation got published in 2015 that concentrated on Finnish  as well as     Estonian candidates[65].In this study a cohort of 33 infants   who had genetic proneness to T1D.Of this cohort ,11 children  generated auto antibodies(i.e seroconverted), as well as     of the11  seroconverted  candidates,4 of them generated  T1D. Microbiome   Evaluation were carried out utilizing 16S rRNA  along with metagenomic  shotgun sequencing data.In total this study observed reduction in  Microbial diversity  as well as      decrease in bacterial gene  amount in auto antibody –positive children during propagation towards T1D. Particularly they observed  a reduction in  Lachnospiraceae as well as     Veillonellaceae in children  generated T1D besides an escalation of Streptococcus,Ruminococcus, as well as     Blautia(figure1)[66].

An external file that holds a picture, illustration, etc.
Object name is fendo-11-00078-g0001.jpg

Legend for   Figure 1

Courtesy  ref no-67-Environmental factors modulate gut microbiota and potentially contribute to T1D onset. Environmental factors, such as birth mode, diet early in life, and use of antibiotics can influence gut microbiota composition and can to lead to lower bacterial diversity, decreased SCFA production and increased gut permeability. Bacterial phyla/genus/species that are affected by environmental factors and differ between T1D patients and healthy controls are depicted in the colors black/green/purple, respectively. Bacterial genus/species that have been identified in proteomic analyses and are increased in either T1D patients or healthy controls are shown in red. Bacterial genus/species that have been identified in metabolomics analyses and are increased in either T1D patients or healthy controls are shown in blue.

A functional  Evaluation observed that bacterial metabolism in auto antibody –positive  candidates had the properties  of a  greater prevalence   of  genes implicated in transport of sugar  as well as     lower prevalence   of  genes implicated in amino acid    generation in contrast to  those that did not have seroconvertion.

In a consequent  DABIMMUNE  study[67],a metagenomic Evaluation of222 Finnish,  Estonian, as well as     Russian  children(with subcohorts of 74  children from every country),that were matched for HLA   risk  as  well as   gender ,observed no association   among islet auto antibodies  or T1D status along with Microbiome composition. Nevertheless,these authors documented variable escalation of Bacteroides species(spp).  Bacteroides species(spp),had lower  excess in Russian  children in contrast to  children from adjacent countries like Finland  as well as     Estonia.They posited  that the greater prevalence    of T1D in Finland   as well as     Estonia were correlated with early life lipopolysaccharide (LPS)  exposure. lipopolysaccharide (LPS)  exposure would potentially  arise from Escherichia Coli (E.Coli)  for Russian  children in contrast to  children from adjacent countries like Finland   as well as     Estonian who would most probably  have early life exposure to Bacteroides species(spp’LPS.

Just like TEDDY, DABIMMUNE  study observed variations in GM composition along with  total reduction in   microbial diversity in T1D subjects.This study further illustrated a potential mode  utilizing ,  in vitro experiments that  explored the structural as well as      functional properties of Bacteroides   dorei  LPS in contrast to  E.Coli  LPS.

2.3Abis Study

All babies in southeast Sweden(ABIS)represents a prospective study that aims to evaluate the birth cohort  for the etiopathogenesis of immune –mediated diseases,in particular  T1D[68].All the mothers belonging to southeast Sweden who delivered a live child between October 1997- October 1999 were requested to take part .Overall equivalent to 17,000 babies  took part in the study (78.6% born in this particular area ), as well as      till date 147  of these went on to generate  T1D.From these newborns blood,stool along with other biological samples were taken at1mth,2.5-3,5-6.8, as well as      11-12 yrs of age .In a study conducted recently from this Abis cohort,the authors evaluated the association among HLA haplotype along with its action on GM  composition[25].Earlier work in mice had revealed ,an inability of presentation of class II antigens/ as well as     /or separate major  histocompatibility   complex (MHC)   class II    haplotypes causes variations in GM composition[25,69].In a study,  the absence of class II antigen  presentation resulted in reduction of lactobacillus spp as well as     escalation of SFB[69].In the  Abis Study ,it was observed by Russell etal.[25],that results akin to mice in humans  having a greater  genetic chances of generation of TID autoimmunity got correlated with  changes in the GM  composition .They realized that the core GM composition     as well as      the beta diversity  varied on the basis  of  HLA   risk group  as well as      total  genotype. Furthermore some protective  HLA  haplotypes got associated  with the genera Intestinbacter as well as     Romboutsia [25].Another intriguing study  evaluated the association  among pets exposure  along with  TID.They observed that  45.5% of pregnant women enrolled  for the study possessed pets .Commonest were cats as well as     dogs ,although exposure to either cats or dogs was not correlated  to TID chances[70]. Nevertheless, they observed exposure to hamsters  significantly enhanced TID chances.This was an intriguing observation since a common study  dependent on  the Canadian  Health Infant Longitudinal Developmental Study(CHILD) demonstrated  that early exposure to household pets  change the GM composition  in the infants  might decrease the chances  of some of diseases like obesity as well as     allergic diseases[71].

3.Influence of environmental factors   on Microbiata GM as well as T1D

In between birth  as well as   age3,the Microbiata of infants  alters at a  dramatic pace  in view of it getting  continuous exposure to innovative environmental /maternal  Microbiata along with food/animal born antigens.By the age 3 ,these oscillations reduce  once the Microbiata   composition  settles  to that of the adult –like situation[60].Early life factors,like mode of delivery ,breast fed or not, cow’s milk exposure as well as     a getting introduced to solid foods have been illustrated to implicate the   early life microbiata composition,thus deciding  its ultimate composition.Hence Early life factors have their impact beyond infancy as well as     a most probably implicate health along withgeneration of diseases later in life .

3.1. Method of Delivery

Earlier work implied that the uterus represents a sterile environment,totally lacking bacteria. Nevertheless, recent research  have queried this.It has been seen in a lot of studies  that at the time of intrauterine  period of growth ,the fetus gets exposure to maternal  Microbiata through transplacental passage into the amniotic fluid[72].Prepartum ,unidirectional transfer in the maternal  GM   among the 1st as well as       3rd trimester  possibly affected by hormonal   along with immunological alterations  at this time period [73].This is believed  to decrease  at this period of time .Intriguingly ,this switch enhances the butyrate generating  taxa which facilitates escalated amounts of immunomodulatory regulatory T(Treg)cells[73].This is  believed  to decrease  the probability of maternal rejection of the fetus[74].A 2nd switch in GM is seen  just before birth.This switch results in greater heterogeneity , decreased alpha diversity , along with over expression  of taxa believed  to stimulate inflammation[74,75].Work conducted in humanized  germ free( GF)  mice has pointed that  these alterations are adaptive as well as      facilitate escalated energy shift  among the mother as well as      fetus [75]. These taxa  are further thought  to facilitate  the proper colonization of  anaerobic spp which are  predominant  in the neonates early life microbiata   .The mother’s vaginal microbiata    also go through changes just before delivery .At the time of pregnancy 4 main Lactobacillus spp escalate markedly as well as      aid in stability  of this community while also decreasing  the    alpha diversity[74].These dominant taxa  don’t have a major part in sustaining vaginal pH or in avoidance of infection,but rather are believed  to escalate secondary  to their significance in original neonatal  colonization[74].

3.1b Delivery Mode as well as part in GM Manipulation

Lot ofstudies have associated delivery mode with clear cut differences in neonatal  microbiata[76,77]. association  among delivery mode   as well as      enhancement  of chances of obesity , asthma,allergic as well as      autoimmune diseases[77-79]. Overall the proof  robustly points  that delivery mode  has a massive influence on   microbiata composition as well as      succession.Those infants that had a vaginal delivery  possess a  microbiome quite  akin to the vaginal microbiome of the mother whereas LSCS delivered babies  get classically colonized  with the species observed on the mothers skin[77].Variations  in the taxa  are maximum separate in the 1st 3 mths of life .Those having  a vaginal delivery  possess  commensal bacteria like   Lactobacillus as well as      Bifidobacterium ,  whereas LSCS delivered babies  are colonized with Clostridium spp as well as      Staphylococcus spp[77]. TEDDY further observed that children Subsequent   to a vaginal delivery   possessed greater Bacteroides   amounts ,that in turn was associated with  a greater   amounts of diversity as well as      rapid maturation of the GM[60].the initial stages of microbiata exposure/colonization  finally decides the composition as well as      succession of the microbiata of the infants along with effects the total metabolism along with immunomodulation .Hence ,initial colonization by abnormal microbiata can cause longterm impact on the immune function along with can enhance the child’s chances of generation autoimmune correlated diseases,like TID.Various studies have observed that  the   colonization of Lactobacillus, Bifidobacterium along with Bacteroides gets delayed or is totally lacking  in infants that are LSCS delivered[76,77,80,81] as  well as      absence of exposure to, Lactobacillus which gets derived from the vaginal that can result in variation in microbiata  succession.Later diversity for the main phyla of GM-Actinobacterium(predominantly Bifidobacterium) Proteobacteria   as  well as       Bacteroides-is further lesser in infants that are LSCS delivered[75,81-83].

3.2 Delivery Mode as well as part in T1D Initiation

In a meta-analysis  conducted by Cardwell et al. contrasted the outcomes of 20 separate studies to find if chances of T1D  associated with Delivery Mode. Following adjustments for covariates like birth weight,gestation period,maternal age , maternal T1D incidence , as  well as       breastfeeding  there was still a 20% escalation  of T1D  initiation in  infants that are lower segment caesarean section (LSCS )delivered[83].Still considering the individual outcomes of studies  shows a lot of discrepancies. Various studies have seen a significant association among Delivery Mode as well as      part in T1D occurrence [84,85], whereas others  have shown little to no link[84,85].

Work regards to a direct association with    delivery mode, microbiota  as well as      T1D incidence is minimal, however proof till date  points that infants that are LSCS delivered as well as      abnormal Microbiota has an impact  on both T1D initiation along with  propagation .In a study involving NOD mouse,investigators  observed   particular  variations  in Microbiota composition   among pups that were  LSCS delivered as well as      those having  a vaginal delivery,though they couldn’t observe a significant correlation among   delivery mode as well as    T1D incidence[86].They further found lesser  amounts of  Foxp3 + regulatory T(Treg)cells,  anti-inflammatory IL-10  as well as      tolerogenic CD103+DCs,pointing that LSCS delivery possesses longterm actions which probably result  in impaired immunosuppression ,  thus escalating  the chances of generation of anti-islet autoimmunity[86].

Further Evaluation of particular genera  that have been believed to Initiate the onset of disease shows dramatic variations in microbiota composition   as well as      irregularities in the part that is played by Bacteroides   spp. Bacteroides   has a significant part in total immune system generation as well as     function by stimulation of plasmocyte generation of secretory IgA[84,87].More particularly ,B.thetaiotaomicron is implicated in sustaining  the gut barrier  whereas B.fragilis as well as      B.subtilis  have been demonstrated   to facilitate  gut correlated lymphoid tissue  maturation  as well as      work in pre immunity   antibody  generation [84,88]. B.fragilis  further inhibited the pro inflammatory cytokine IL-17 in the intestine [84,89].

On the other hand  one of the maximum observed differences  in microbiota for the seroconverted high risk children  was the escalated  Bacteroides  amounts. An example is the longitudinal   Type 1 diabetes mellitus Prediction as  well as     Prevention( DIPP) Study,which is a Finnish research work that Initiated blood, along with stool samples from children possessing   high risk HLAgroup genotype in 1994[90].A study  that was more recent,  utilized     4 matched case  control  pairs  from DIPP  to isolate  Microbiota   that varied among   cases  as well as controls[91].Depending on their Evaluation,one main difference  was the excess of Bacteroides as well as  Firmicutes . Cases  possessed a significantly greater   escalation of  Bacteroides,that  enhanced   over time in contrast to  controls .Validating these outcomes ,of the taxa  isolsted in the DABIMMUNE  study , Bacteroides spp were observed to be  maximum in excess in the seroconverted  cohorts from Finland   as well as     Estonia[91]. Studies akin to this  observed that Bacteroides spp got overexpressed  in case subjects, particularly the spp Bacteroides   ovatus ,that was implicated for 24%of the escalation found[91].A study that was more current Finnish  one[92] validates these observations utilizing 76 at high risk children  that they monitored  from birth to 2 yrs  of age.Metagenomic Evaluation isolated 2 spp  B. dorei as  well as       B.vulgatus ,which were significantly  greater in cases in contrast to  controls before seroconversion[92]. Nevertheless, a lot of Studies have arrived at separate conclusions  or have observed no association  at all among Bacteroides escalation  as well as     T1D Initiation[93].Figure 1 shows a summary of these observations  as well as     the variations  in the microbiata  composition  of T1D patients along with controls.

3.4 Role of Breast feeding

Lot of work has observed  the advantages that Breast milk yields for an infants growth as well as     generation[94-96.Bioactive agents possessed by Breast milk are antimicrobial  as well as     immunomodulatory components ,have got demonstrated to modulate the GIT as well as     immune  function,beside GM composition via multiple modes of action.[94-96].

3.4b Role of Breast feeding   in Microbiota  Manipulation

Besides Bioactive agents,the maternal  microbiata  gets shifted in Breast milk as well as     alters  infants GM that are associated with the  rate  of breast feeding in a dose-bases way[97]. Particularly ,lactic acid bacteria  in the genera Bifidobacterium as  well as   Lactobacillus,possessing the ability of  breaking   down  human milk oligosaccharides(HMOs),get shifted from the mother towards the child[98].These genera  are believed to sustain the intestinal  barrier ,induce the generation of IgA antibodies   as well as  gets    implicated  in the    generation of short chain fatty acids( SCFAS)([99-102].Noticably ,the spp Bifidobacterium longum subspp infantis gets excessive in infants   which received only breast feeding   strictly for1st 6mth of life[60].B. infantis is  Specifically efficacious in the metabolism of HMOs into SCFAS(indirectly) along with has the ability  to facilitate mucus generation,ameliorate diet induced colonic mucus degradation  along with  possess significant part in immunomodulation[103](figure1).

3.4c Role of Breast feeding   as well as part in T1D Initiation

For the  immunologic generation successfully  crosstalk among host along with its Microbiota at the mucosal surface of the intestine  needs to occur [102,104].In murine studies  key generational windows  where  microbe –driven immune-control can take place .Similarly in humans it holds true –detailed later in gluten dependent foods .Thus  these results point that the existence of   Microbiota along with the time of these microbiota get introduced  are crucial  for the appropriate immune system generation.For those children who have genetic proneness to T1D,  Breast milk is believed  to possess protective characteristics against T1Dgeneration[105].Furthermore ,infants  who are only receiving Breast feeding    during the initial 6mths of life  have a separate microbiota composition  .It has been posited  that these protective characteristics of Breast milk  are efficacious by manipulation of GM composition[106-108].

In 2015 ,MIDIA,a Norwegian study   Evaluating the correlation among Breast feeding    time period, age at which solid foods get introduced , as well as     the chances of islet –autoimmunity/ T1D in  children possessing genetic proneness to T1D observed  that any particular frequency of Breast feeding     for12mths  or greater had an association with    a slower propagation  towards  as  well as   total reduction in T1D incidence [109]. Nevertheless,  both age of solid foods introduction,or if infants were getting  Breast feeding     at the time of introduction were seen to have any action on islet –autoimmunity as well as     or T1D propagation[109].These outcomes point that    Breast feeding     by itself possesses  having a significant influence on T1D propagation/ Initiation,whereas the introduction of other food sources ,like solid foods ,might not have a significant influence[110,111].

Most of the studies conducted  have concentrated on the Firmicutes :Bacteroides ratio  that is a potential indicator  for the generation of disease.Despite  the association among  disease along with switches in  greater ratios is just a posit ,similar  switches in  Bifidobacterium along with Bacteroides  have been seen in children having a chance for T1D generation.As both genera have the ability of metabolizing  HMOs,this inverse association is believed to originate  from inter particular competition  regarding the same source ]112].Intriguingly  these switches get parallel in contrast amongst  Breast fed  (greater Bifidobacterium) as well as    formula fed(greater Bacteroides)infants.It is not astonishing ,an inverse association among Bifidobacterium  colonization  as well as     T1Ddiseasegeneration has been seen  in a lot of cross sectional along with longitudinal  human studies[102,112].Furthermore Meija-Leon etal. recognized  an  escalated population of Bacteroides   has been illustrated  in recently diagnosed T1D[113].Akin to that a greater T1D incidence in Finnish as well as     Estonia study   population has been seen   along with a  greater prevalence of  Bacteroides in at risk children in contrast to   children at risk in adjacent Russia[61].

In the other DABIMMUNE  study detailed earlier ,authors demonstrated that Bacteroides spp,like       B. dorei,    inhibited  immune stimulation as well as     inflammatory cytokine  responses to  E.Coli  that lead to repressed innate    immune signalling as well as     reduced endotoxin tolerance [112].This immune  repression gets manipulated  by high amounts of Bacteroides-obtained  LPS which are structurally as well as      functionally  separate  from those generated  by E.Coli  ,the dominant kinds of LPS existing in Russian infants [112].This immune  repression  gets mediated Furthermore in vitro  experiments demonstrate that    LPS  generated by different taxa  can inhibit or stimulate  toll like receptors(TLRs),TLR4, NFκB  activation as well as     endotoxin tolerance[161-67]. Particularly, when  NOD mice got injected with an immunogenic LPS  generated  by E.Coli , endotoxin tolerance was evoked    In  addition to    reduction in T1D incidence[112]. LPS from B. dorei, did not confer similar protection against T1D Initiation[112].These outcomes  validate earlier NOD mouse studies where LPS was observed to have a direct  effect on T1D propagation[112,115].The mode implicated  in LPS  manipulated immunity as well as     its association with    T1D generation are not clear totally ,but earlier work by Guiden etal.[116], as well as     Wen etal.[55] have demonstrated   the significance of toll like receptors(TLRs),TLR 3 as well as     innate    immune   signal transduction  Adaptor(MyD 88) in T1D Initiation in NOD mice  ,that is 2 parts of the LPS/ TLR4 signal transduction   pathway[55,112,116].

Together these outcomes from both in vivo along with human studies  have found the significance of some Breast milk  taxa in immune system  generation along with manipulation.Certain genera,like  Bifidobacterium   have a significant  part in the total health  as well as     generation  of an infant , as well as     have further been illustrated  to  have a particular,    part  in conferring protection  to children  at risk  of T1D Initiation(fig1). Nevertheless, in current studies,even those infants  who were only receiving Breast feeding    during the initial 6mths of life   had no Bifidobacterium spp, pointing  that   mothers are not getting  colonized  by these     genera to the same degree  as their predecessors.In a study from USA ,30% of the Breast feeding  infants had no Bifidobacterium that could get detected , as well as     for those infants getting Bifidobacterium   colonization  ,only 30%possessed Microbiota in which Bifidobacterium accounted for greater than 50% of   the   population [102,117].

3.5 Role of   Dietary  Factors

The total T1D incidence has  escalated significantly  in the last half of the twentieth century[117,118].Existing proof has suggested that Dietary  factors aid in T1D Initiation.Certain factors  have  pointed  to stimulate  or  escalate propagation of disease,whereas others  confer protection against the  generation of     T1D associated  autoantibodies along with total propagation of disease[59.117,119-125].Animal along with human studies have pointed  that   early life  exposure to foreign food  antigens,like gluten  as  well as     bovine insulin can impact   β cell    autoimmunity[126]. Nevertheless,exactly the way along with the degree  these Dietary  factors influence disease results still has to be found

3.5a Gluten-Dependent Foods

A usual physiological property of T1D etiopathogenesis is an intestinal barrier that has become weak (alias leaky gut syndrome) that aids in escalated inflammation  in T1D patients[34,127]. Gluten- is made up basically of monomeric  gliadin  along with polymeric  glutenins  that can further aid  in escalating gut permeability  as  well as        stimulate inflammation by cytokine liberation[128].Hence intestinal barrier function are key  for suppression of inflammatory response . Gluten-Dependent   diets are believed to aid in gut permeability via the manipulation of GM[129].Validating this taxa correlated with a Gluten-Dependent   diets,like Akkermansia spp have been demonstrated to control human tight junction proteins,confer protection  from pathogen invasion besides having a protective part against T1D initiation[130].

3.5c T1D initiation as well as Gluten-Dependent Foods

Till now the results on action of gluten-Dependent  Foods have demonstrated lot of variability.A lot of studies have observed that putting  patients  on  gluten -dependent  Foods does not improve their antibody  titres to a great extent . Nevertheless,  same studies have further   demonstrated that  gluten-Dependent  Foods can improve a person’s GTT[131].Few studies concentrated on  patients with both celiac disease as  well as        T1D found a significant  advantage with respect to improving health as  well as        DM regulation  in those patients receiving gluten-dependent  Foods[132]. Nevertheless, other studies have not observed akin actions[133].

Inspite of these inconsistencies ,1 factor has been commonly implicated  as having a key part  in generation of disease  namely the time along with method of gluten administration.Human studies observed that early exposure (<3mths of age) to  Gluten escalates the risk of islet auto immunity as  well as        this action gets ameliorated  if Gluten is delivered when the child is still  being given Breast feeding   [43,134].Similar actions are observed in animal studies[135], as  well as        has been  found to impact the initiation of T1D in a dose –based way[136].

The diet of the mother at the pregnancy time was evaluated  in association  with a child’s chances of T1D generation.On feeding the NOD mice   mothers  a Gluten-free  Foods,the T1D incidence in their offspring got significantly decreased [137].In a study akin to this astonishing 7  times reduction in the T1D incidence in NOD mice   pups occurred(62.5%-8.3%)[137]. Nevertheless, the outcomes from human studies have not been consistent.A current study by Antvorskov  etal.,[138]observed  that a cohort of Danish women,the risk of generation of  T1D was directly proportional to how much gluten was consumed during pregnancy.,[138].Women having the maximum intake of Gluten(>20g/day) possessed double the chances of T1D initiation in their children. As   compared to that,2 previous  studies observed no robust chances  among mothers   as  well as   T1D incidence in the child[139].

Getting exposed in early life  to diabetic Dietary  factors  possesses main impact on T1D initiation.Inspite of that deletion of Gluten from diet  of older children  having a diagnosis of T1D has been illustrated to  enhance diabetic metrices  that are propagation of  remission periods  along with decreased HBA1c[140].A study akin to that documented   better glucose tolerance  as  well as   insulin sensitivities ,but no advantageous action in relation to better titres of islet auto antibodies in older children  put on Gluten-free  foods for6mths(median age=16yrold)[131].

In total these research works  have given key knowledge on the part gluten  plays in T1D initiation. Nevertheless, human longitudinal studies are required to derive an association with diet, its impact on the   Gut Microbiome  , as  well as   how this influences T1D initiation as  well as   propagation.1 such study is the German longitudinal   cohort study, BABYDIET study.

Babydiet Study

BABYDIET represents a substudy of the bigger BABYDIAB study that got started  to find if gluten G intake  in early life   has a part in T1D initiation.This study  had been following 22 children showing auto immunity as  well as   22 matched control for slightly greater than 3yrs .They had stool samples procured mthly  among ages 3mth-36mths along with 6mths interval following that .All the participants had a minimal of one first degree relative  having overt T1D.In toto no significant variations were seen among cases as  well as     controls  for the Microbial diversity (namely rich or evenness) along with  composition once results had been corrected  a lot of comparisons[141]. Nevertheless, on contrasting the total Microbiota network (Eigenvector Centrality (EC), Node Degree] of children showing autoimmunity vis a vis autoantibody  negative children, significant differences  were observed . Autoantibody positive children illustrated significantly   separate centrality evenness in contrast to healthy controls at 6mth along with 2yrs  of age .Further greater  network nodes  having intermediate  connectivity  in the autoantibody  positive children cohort. Moreover, particular genera  had differences   in cases  along with control  in an age-based method.Like at 6mth of age ,the bacterial genera  Enterococcus ,Prevotella , as  well as   Corynebacterium displayed   greater  EC at 2yrs of age in autoantibody  positive children. As   compared  to that genera  which documented  greater  EC at 2yrs of age in autoantibody  positive children had Barnsiella as  well as   Candidatus Nardonella ,while  Staphylococcus  as  well as   Nocardioides  possessed greater  EC in the autoantibody  negative children.It is significant to notice  that these findings  were at the genera level  secondary to 16S sequencing.

3.5d Cows Milk Along with Its Proteins

Both animal model studies along with   human ones have observed correlation among  T1D incidence along  with the intake of Cows Milk at both weaning time as  well as   later in generation.Retrospective  meta-analysis  on national dietary intake records  have got utilized  to find  if national along with global  standards of Cows Milk administration  associates  with  regional     T1D incidence rates[126,142,143].Various Evaluation  conducted by Montoni etal.,observed a positive correlation among T1D along with an areas  consumption of animal –dependent energy  as  well as   the areas total milk supply [217].  A meta-analysis     that particularly contrasted a countries T1D incidence  rate with its yearly Cows Milk  protein intake also observed a  positive  association with a countries T1D incidence  rate with its yearly Cows Milk  protein intake   [126,143].A negative correlation among breast feeding     (at 3mths) as  well as   T1D chances was further seen [126].

Type 1 diabetes mellitus Prediction as  well as     Prevention( DIPP Study),a Finnish Study evaluated usual food intake  in children possessing   extensive β cell autoimmunity along with  observed  that the usual input of  Cows Milk products appeared to be   one of the rare factors  that had direct  correlation with  β cell autoimmunity.This was not seen with other dairy products ,like sour milk products like cheese ,that points  that  the enhanced chances of     β cell autoimmunity arrives particularly from Cows Milk  correlated proteins [143].Various Studies have  evaluated   the immune response to these proteins as  well as   observed  escalated risk of Cows Milk  particular IgG as  well as     IgA antibodies at the early T1D initiation[145].Intriguingly ,Elliot  etal.,[143]observed that the total  proteins intake did not associate  with      Type 1 diabetes incidence  as the particular intake of Cows Milk  -obtained β-casein(A1 variant) did[143].These findings  point that     the escalated humoral response gets exhibited in against Cows Milk  proteins in T1D patients,that makes them a potential aetiological factor resulting in the autoimmune event  leading to T1D initiation[145].

Whereas proof agrees with a part for Cows Milk  proteins in T1D initiation,other studies have not observed any such association[146].On the other hand,other  studies have pointed  that   Cows Milk   dependent formula  has a protective part against    T1D initiation[147].Human studies , evaluating  the association  among Cows Milk  proteins ,GM composition as  well as   T1D have been obtained from 2 main longitudinal  human studies.i) Trial to Reduce IDDM  in the Genetically at Risk (TRIGR ) as  well as   ii) Finnish Dietary Intervention Trial  for the Prevention of type 1 diabetes (FINDIA). Whereas  these studies do not particularly look at the correlation among Gut Microbiome  along with  T1D incidence,a crosssectional study obtained from these studies  was later carried out to evaluate these actions.

 3.5di) Trial to Reduce IDDMin the Genetically at Risk (TRIGR)

In this TRIGR  study,external dietary proteins got postponed till 6-8mths of age as  well as   the generation of autoantibodies(insulin, GAD,IA2 ,ZnT8) was screened till the age of 6yrs[119].The objective of this study was to  if adding  breast milk with greatly hydrolyzed milk formula had the capacity of inhibition of T1D initiation.Infants possessing verified   HLA-correlated  proneness(n=230) along with a minimum of 1 family member having T1D recruited in the study.These patients were delivered either casein hydrolyzate  formula or conventional formula wherever breast milk was not present as  well as   were later followed for till the age of ten.During this study ,17  children  generated  at lease one Autoantibody in the casein hydrolyzate   group(17%) in contrast to  33 children belonging to the control group(30%).8 children in the casein hydrolyzate   group(8%) as  well as   ,17  (16%)   in the control group generated  2 0r > autoantibodies.Insulin  autoantibodies were the commonest  seen  first – autoantibodies with anti-islet autoantibodies remaining a close 2nd .In total this study observed weak , correlation   among formula introduction along with  with an escalated generation of T1D. A  greater current update of  this   study got published in 2018 observed that weaning  to hydrolyzed milk formula did not decrease the chances of  generation of T1D with an escalated   disease chances upto a median age of  11.5 yrs[148].

3.5dii) Finnish Dietary Intervention Trial  for the Prevention of type 1 diabetes (FINDIA)

FINDIA represents a multisite double blind –clinical trial where newborn infants got randomized  to get either bovine insulin  free cow’s milk formula(CMF, CMF group),a whey dependent hydrolyzed  formula(WHF group),or a whey dependent   FINDIA formula (FINDIA  group)from where bovine insulin   had been deleted .Among 2002-2005, subjects got enrolled from 3 Paediatric hospitals  in Finland.113  infants with HLA-correlated  proneness to T1D were randomly allocated to get one of the three formulas .These outcomes from the FINDIA study [149]point that delivering bovine insulin-free    formula at the time of 1st 6mths of life  decreases a child’s chances of generating β-cell autoantibodies    by the age of 3 .Once the Microbiota gets seroconverted   subjects got contrasted with non seroconverted   subjects,an  escalated  amount of Bacteroides, along with reduced amount of Bifidobacterium.   Evaluation of  intention to treat ,6.3% of children in the CMF group,4.9% in the WHF group, as  well as   2.6% of children in the FINDIA formula group   were positive for a minimum of 1 autoantibody by the age of 3.These scientists observed  that in contrast to   ordinary formula, weaning to an insulin free formula (TRIGR group)diminished to cumulative  incidence of    autoantibodies by age 3yr in children at genetic risk of T1D.

3.5diii) TRIGR-FINDIA Cross-sectional study

In a study by De Goffau etal.,[32]GM composition  was evaluated  along with contrasted with autoantibody positive(n=18),who tested positive for a minimum of 2 autoantibodies) along with matched autoantibody negative –negative (n=18), children.These candidates got enrolled from TRIGR, as  well as   –FINDIA. in contrast to    autoantibody negative  children, autoantibody positive children possessed relatively greater amount of Bacteroides(Bacteroides genus). Nevertheless, at the species level only little Bacteroides spp were correlated with  auto immunity, along  with a greater degree of Firmicutes, like  Clostridium perfingens,both of which are agreed to escalate  gut permeability.This Evaluation further points that   low amounts of    Bifidobacterium adolescentis  along with Bifidobacterium pseudocatenalutum(<12% in combination) have a   significant association with   escalated amounts of  β cell auto immunity (figure1) .

3.6Antibiotics

Antibiotics get commonly utilized   for the regulation of bacterial  antigens, possessing a wide variety of actions on the GM.Exposure to  antibiotics has been observed to diminish  the bacterial diversity significantly, Nevertheless,various studies have demonstrated that adult GM usually recovers to simulate  Microbiota before treatment[150].However, A 4day treatment with a mixture of 3  Antibiotics(meropenem,gentamicin as well as      a vancomycin)resulted in the deletion of 9 bacterial   spin adult men 180days Subsequent to antibiotics use[150].In the same study it was seen that an  initial flourish of pathobionts  like  Enterococcus  faecalis as well as     Fusobacterium nucleatum along with  a reduction of Bifidobacterium[150].

Usually on average basis ,a child in the USA receives 3 Antibiotics courses  prior to the age 10[151].Inspite of common delivery  to infants as well as       children ,their actions on GM along withhow it associates  with  human diseases,like   T1D is not totally clear.A   study of  infants , Exposure to  antibiotics was correlated with  reduced  Clostridiales as well as      Ruminococcus in the initial 3-9mths of life , along with postponed maturation of the  microbiota[152].Conversely ,in a large Norwegian Mother along with Daughter  Cohort study did not observe any correlation with acetaminophen  utilization  along with T1D chances.In this same study  utilization of antibiotics was not associated  with greater  T1D chances   in both Mother along with Daughter in the 1st 6-9mths  of life[153].Akin to that TEDDY observed that frequency as well as       utilization  of antibiotics in the initial 4yrs of life had no impact  on the chances of generation of   Autoimmunity for T1D or Coeliac disease[154].A case  control  study that was population-dependent as well as       included all T1D children born from 1997 did not observe any association  among utilization  of antibiotics   along  with  generation of     T1D[155].In a population-dependent mother child cohort  which had all children born from 1996-2000 in Finland,that got a diagnosis of T1D,an escalated risk of generation of   T1D was observed in children born to mothers consuming phenoxymethyl penicillins or quinolone prior to pregnancy.They posited that  utilization  of antibiotics by Mother’s might reduce  the shifting of healthy microflora to the baby.An  escalated risk of T1D was also  observed when the mother’s consumed macrolides   prior to pregnancy as well as       child received macrolides  in contrast to   mothers-child pairs when none of them received macrolides.  A greater risk of antibiotics ,as the definition said ,namely 7 or  greater buying of antibiotics was also observed to have an  escalated risk  of generation of     T1D. Bifidobacterium   as well as       Lactobacillus spp get correlated with a healthy microflora,being Specifically sensitive to macrolides, quinolones  as well as       penicillins.This study posited that macrolides, along with quinolones  might  escalate the  risk of   T1D in children by avoiding the generation DNA along with the enzymes in β cells leading to β cells demise[156].  Another study  that was case  controlled  utilized    THIN,i.e ,a UK population medical record database ,which has total medical records of  approximately 10 million patients,for evaluation of the action of  antibiotics exposure on both type1 as well as         type2 diabetes  mellitus.The study observed  that evaluation of single antibiotic was not correlated with  greater  diabetes  chance. Nevertheless,receiving 2-5 courses of penicillins, cephalosporins, macrolides,or  quinolones   had a   correlation with  greater  diabetes  chance, as well as       intake of over 5 courses of  quinolones was correlated with  greater  diabetes  chance.  Getting   exposed to greater than 5 courses of  penicillins further was association with    greater  diabetes  risk[157].

Various studies  have got conducted on biobreeding diabetes prone (BB-DP) rats ,LEW1.WR1 rats along with NOD mice to find out the  influence of antibiotics therapy on T1D incidence as well as       Gut Microbiome composition.Brugman et al.[158], studied male along with female BB-DP rats on a conventional(CON)plant dependent  or hydrolyzed casein(HC)in continuity got antibiotics therapy with broad spectrum antibiotics Bactrimel(sulfamethoxazole as well as       trimethoprim) along with colistin  sulfate. antibiotics therapy when on  the CON diet  was observed to decrease T1D incidence as well as      postpone  the initiation to 30days ,while antibiotics therapy when on  the HC  diet  was observed to confer protection  against initiation of T1D[158].Further BB-DP) rats who generated T1D later  were illustrated To have  escalated amounts of Bacteroides in contrast to  controls  as well as       rats which did not  have T1D generation[158].Akin to this ,in 2012,Hara et al., Evaluated LEW1.WR1 rats that were infected with kilham rat viruses (KRV) for induction of T1D, observed  that virus stimulated   T1D or repressed  by treating with  antibiotic sulfatrim .Therapy with sulfatrim was further  observed  to repress the virus stimulated   anti-islet responses  in the LEW1.WR1 rats through down modulation of immune system.Moreover , infection with KRV viruses  resulted in Bifidobacterium   along with   Clostridium escalation[159].On the other hand ,Livanos et al., observed that pulses of therapeutic dosage of the macrolide antibiotic tylosin tartrate(pulsed therapeutic antibiotics ,PAT) exaggerated T1D as well as       insulitis generation in male NOD mice.The study further illustrated that the group that  received  PAT had greater amounts of Akkermansia along withEnterococcus  in contrast to  control groups  that had greater amounts of Bifidobacterium[160].Hansen et al.,[161] conducted a study in 2012 that evaluated the early life  therapy with  antibiotic  vancomycin  on  NOD mice.They illustrated that mice receiving  vancomycin from birth till day 28 possessed a significantly lesser  T1D incidence as well as       greater clusters of CD4+T cells generating  pro inflammatory cytokines.On the other hand , NOD mice who got  vancomycin from age 8wks  till T1D initiation possessed  greater T1D incidence, as well as   greater insulitis score    along with  escalated glucose amounts,that  pointed that    early life treatment significantly  influences the   T1D propagation.Further  vancomycin treatment also influenced the  Microbiota composition  since Akkermansia  muciniphilia  was dominant in the Gut microbiome  of all groups  receiving   vancomycin]161].In a  study  akin to that ,the actions of   antibiotic therapy on  T1D propagation in the offspring of  NOD mice  getting antibiotic therapy  during pregnancy  further revealed  the significance  of antibiotic therapy  in early life[162].These authors gave  during pregnancy   a neomycin,polymixin B as well as         sptreptomycin ,ie 3   antibiotic s  to the NOD mice    along  with illustratedthat offsprings of mice  getting antibiotic therapy had   significantly    greater protection conferred in contrast to  mice who got antibiotic therapy immediately following birth  as  well as     mice that NOD mice  getting antibiotic therapy   at 3wks of age.This study highlights  the  significance  of antibiotic therapy  in early life or prenatal  antibiotic therapy  to counter T1D since the age of mice  getting treatment possessed a negative association with protection from type1 diabetes.  Moreover, the antibiotic therapy   was observed to remove gram negative Proteobacteria as well as          result in gram positive bacteria predominantly belonging to the Firmicutes  family. As per the studies documented,it is not clear  if antibiotic therapy    constantly escalates or decreases  the type1 diabetes risk  in animals,but it is always observed to impact the microbiota composition   as well as          T1D incidence along with proof to point that   early tackling is more efficacious.

4.Type1 Diabetes along with GM:peeping beyond the bacterial species

Once sequencing got introduced ,it changed the manner by which researchers  asked queries regards to both environmental as well as          human correlated microbiota ,thus has made them utilize big data  in a manner that was never feasible earlier.With the advancements of sequencing methods , greater researchers  are trying to utilze these technologies  for Evaluation of correlation among the human Microbiome along with autoimmune  diseases.At present , type1 diabetes  research, researchers have Evaluated the variations  existing in gut proteome,virome along with metabolome  of T1D patients.Here the advances that have been conducted in the omics field is detailed.

4.1 Gut Proteome

16S along with shotgun sequences studies give us information  as far as the Microbiota composition   of the Gut Microbiome is concerned .Whereas Proteomic studies are required to get an insight  regards to the proteins manufactured by the gut microbiota.Occasional studies till date  have got carried out  for Evaluation of  intestinal  Microbiota  Proteome of Type1 Diabetes  patients. Nevertheless, in a study Pinto etal.,[162], Evaluated the variations in the intestinal  microbiota  Proteome among children with Type1 Diabetes   that had got established(n=3) in contrast to   Proteome  of healthy children  ((n=3).In control samples , bacterial proteinsfrom Bifidobacterium adolescentis,  Bifidobacterium longum subspp infantis,Ruminococcus,Collinsella  aerofaciens,Coproconus comes as well as          Clostridium spp were  observed to be the maximum enriched,whereas proteinsthat initiate from Eubacterialis rectales,Faecalibacterium Prausnitzii, Bacteroides dorei   along with Bacteroidesuniformis  differed among control as well as      case  samples(figure1).More currently ,in one study[163]stool samples from a newly originated  Type1 Diabetes patients,islet- autoantibody positive  along with low risksubjects were contrasted.

MetaProteomic  Evaluation got used for differentiation of  stool samples   into 3   divisions depending on the existence of human(host-originated) along with microbiota  originated proteins.In case of  patients from   newly originated  Type1 Diabetes ,a significant decrease in microbial-correlated host proteins ,which sustain the  mucus barrier,adhesion of microvilli  as well as        exocrine pancreas .Hence  Type1 Diabetes patients  possessed  a greater prevalence of intestinal  inflammation  along with  reduced barrier function.An earlier study  that got published in 2016 laid the platform for large scale metagenomics  studies  by illustrating  on a smaller level ,the way particular gastrointestinal Microbial communities  as well as         host particular

phenotypic association are linked to initiation of human diseases[22].Though the part of exocrine pancreas  In T1D initiation is not clarified  they illustrated that exocrine pancreas  enzymes like amylase,carboxypeptidaseCPA1 as well as  CUZD1 are lesser in  amount in Type1 Diabetes patients[22].These early outcome  seem to be attractive along with giving marked understanding regarding how T1D MetaProteome varies  in function in contrast to  controls .More research in this field  will give greater knowledge  in the propagation of disease as well as        avoidance methodology.

4.2 Gut Virome

Viruses  have been believed as potential  stimulators of T1D initiation  along with propagation[165]. Nevertheless,most of the research has concentrated on the part of viral infections  in this event   as well as        co conclusions have been constructed out of that  data .More  currently   researchers have evaluated thepart of the Gut virome.This Gut virome Is markedly    less explored  since it associates both states of health along with disease , Nevertheless, the properties of intestinal virome from birth towards the generation   of  autoimmunity possesses the capacity to become a significant part in getting insight  in the etiopathogenesis of T1D. Utilizing the longitudinal   Type 1 diabetes mellitus Prediction as  well as     Prevention( DIPP)cohorts, from a Fnnish Study,stool Viromes got acquired from 19cases(who became autoantibody positive prior to the  age of 2 as well as    later  generated T1D along with  matched controls at 0,3 as well as       6mths prior to the   initiation  of islet autoimmunity[165]. Viruses comprised 2% of all the sequences that were isolated ,having a main dominance of bacteriophages  that got isolated in 52/96 samples(54.2%).Astonishingly ,only 10.4 % of samples possessed  1 or greater human Viruses like parechVirus,boca Virus,annelo Virus,entero Virus as well as       /or sapo Virus.Kramma etal.[165],further observed no correlation among the Gut virome as well as        islet autoimmunity[166].

A study akin to this was carried out in 2017[167], Evaluated stool samplesfrom 11 childrenwho had generation of autoantibodies in correlation with T1D(of these 5 had T1D generation). in contrast to  Kramma etals[166] .,study this one observed a lesser amount of Circoviridae –associated sequences (this group of viruses  included circovirus) as well as lesser virome diversity in cases of T1D.More significantly the workers found that enough proof was there regarding with time ,the generational propagation  of viromes  was significantly separate  among the children  who   had autoimmunity  generation from those who did not.The dynamic alteration in Shannon diversity  with time  was separate among controls  as well as        T1D cases for Microviridae T1D as well as        Podoviridae.The Random Forest  evaluation of age –discriminative contigs further pointed that  there was variation among ` case  as well as       controls [167].This study   illustrated viromes alteration  took place prior to seroconversion  that points to probable causality.Utilizing virome capture sequencing  techniques a more current study  evaluated the gut virome of pregnant women  with (n=35) along with without(n=26)TID right through the Environmental Determinants of Islet  Autoimmunity longitudinal studies.Two viruses ,picorbina virus  as well as        tobamovirus,were found with greater frequency in pregnant women with T1D,as well as  the enrichment of 77 viruses was separate among the 2 maternal groups  that included 88 enterovirus B kinds[168].

In toto, these results point that    variations in the gut virome of  T1D cases  as well as propagators. Nevertheless,one needs to use a little cautious in trying to decipher these observations as very occasional studies  utilizing a markedly small sample size have been carried out on this topic.Like in other new evaluations,variations in techniques  among studies might result in the differences seen in the conclusions.

4.3 Gut Metabolome

In vivo studies have illustrated the significance of  GM in the generation of host metabolites [169,reviewed by us ref 6].Of the metabolites known to get manipulated SCFA, Specifically have been illustrated to have important part in many respects regarding homeostasisin general.On intake of dietary fibres,it gets digested  followed by  fermentation in colon by GM into short chain fatty acids( SCFAS)(like butyrate or butyric acid  (BA),acetate(AC) as well as  propionate  [169].Diets that possess high fibres change GM composition by promotion of the generation of SCFA –generating spp.Moreover a 4 times escalation in SCFA has been seen in breast fed infants in contrast to   formula fed ones[170].

Further Diets that possess high fibres also have been demonstrated to decrease systemic inflammation[168].A study conducted by Trompette etal.,[171] showed that mice receiving these diets high in fiber  possessed greater amounts of SCFA  along with got protected from allergic inflammation  of the lung [171].In case of  human beings a reduction in   diets fibres  has got directly correlated with a reduction in  SCFA- butyrate [171].This butyrate  is the particular SCFA possessing  great significance.Of its lots of parts ,it enhances glucose  along with  energy homeostasis ,acts as an energy substrate for the colonic epithelium  along with represses colonic inflammation  as well as     carcinogenesis.Moreober   generating spp.Moreover butyrate   has a necessary part in manipulating the immune responseby stimulation of differentiation of regulatory T(Treg)cells, SpecificallyFox3p+ Tregs as well as     inhibition of inflammatory cytokines,like IFNƴ[27,169,173].Intriguingly on  feeding NOD mice,a diet that has been fashioned to escalate  acetate  as well as       fbutyrate or butyric acid  (BA),amounts ,they had a reduction in   T1D incidence in contrast to  controls[174].

In 2 longitudinal studies like TRIGR along with –FINDIA  , SCFA generating spp like Bifidobacterium Adolescentis,Roseburia faecis, as well as     Faecalibacterium Prausnitzii had a negative association with     the  autoantibodies amounts observed[32].Hence children who had 3 or greater autoantibodies had the least amounts of SCFA generating spp.The TEDDYstudy validated this point  finding a greater prevalence of Microbial genes  correlated with fermentation  as well as     SCFA  generation in healthy controls.Intriguingly ,the taxon correlated with these functions  , were not  persistent as per the geographical area   pointing  that     the function  instead of  the taxon is conserved in healthy children[19].Overall,these outcomes point that    spp having the ability for generation of  butyrate have the ability to confer protection  against autoantibodies generation in at –risk children, more particularly different Clostridium clusters  having the ability of generation of butyrate from acetate , along with Bifidobacterium spp that can result in generation of butyrate through lactate metabolism(figure1).

Akin to the observations  seen in animal models ,human studies have illustrated a metabolic  switch in patients prior to the   autoantibody generation,insulitis , as well as    /or T1D.Sardinia  possesses  the 2nd biggest incidence just following Finland,thus is a good  area for T1D studies.In a Sardinia-dependent study , Culeddu etal.,[175] observed that gut microbial products,like p-cresol sulphate along with phenylacetylglycine  were implicated in the  grouping of T1D patients . Utilizing results obtained via the  Finland-dependent  DIPP   longitudinal study, serum metabolomic profiles of children propagating to T1D got contrasted to    controls not  possessing  autoantibodies as well as    /or had generated T1D.For the subjects who had propagated  to T1D,decreased amounts of succinic acid as well as     phosphatidyl choline (PC)were seen at birth,whereas decreased amounts of  triglycerides  as well as     antioxidants  ether phospholipids  were visualized in serum samples .Escalated amounts of  pro inflammatorylyso PC’s , decreased amounts of  ketoleucine along with escalated  glutamic acid  were the changes  observed months  prior to the seroconversion to autoantibody positivity [177]. Intriguingly, Metabolic profiles  became partially normal  for T1D patients following seroconversion.This points that autoimmunity presents later  following  an earlier change  that might have impairment in Metabolic  function[177].

5. Conclusions

Thus lot of work has been carried out  to get total insight  into the etiopathogenesis of T1D,how it originates along with propagates..One thing is apparent ,that Type1 Diabetes is not simply secondary to a single etiology but implicates multiple factors.Moreover the techniques utilized for evaluation of this complicated disorder might have an impact on the results seen.One big variation stems from the conclusions derived from animal studies vis a vis human ones.Despite animal studies giving us a lot of deep knowledge regards to the particular mode  working ,lot of variations  are there inherent to rat/mouse  models  make one  use these data with a pinch of salt as far as human health applications are in question.Like histological  reports have illustrated definitive variations  regards to both the physiology of pancreas  in mouse models along with propagation   regards to etiopathogenesisof T1D in the β-cell islets[178].Furthermore epidemiological results on T1D documented a little male preponderance of the incidence rates prior to the puberty, nevertheless,the incidence is 2 times greater in  male in contrast to  females amongst the ages of 15-39[179].In case of mice female NOD mice generate T1D at a more  greater speed in contrast to   the male ones.There was one study that revealed a lot of Bacteroides   genus  associations were sex-based .A particular e.g is the B.ovatis that was observed to have  a positive association with autoantibody positive male children whereas the same spp had a negative association with femaleautoantibody positive persons[32].Hence future study into these apparently  elusive variations  is required  to find if sex variations are actually  secondary to inherent variations in the GM.It is significant to  know that scientists have isolated approximately 500 separate treatments to avoid or correct T1D   in NOD mice models ,where neither of these therapies got translated  into an efficacious  method for human T1D   [180].The   variations  amongst NOD mice model as well as     human results  point that    one has to consider other animal models for research in T1D  as well as     concentrate on human  studies.

With the advancesin sequencing method newer avenues for evaluation of human  microbiota  have got available in biomedical research.The part of microbiota regards to a lot of chronic life implicating diseases can be studied including T1D.Conversely till now maximum approaches have been bacteria centred.Very little virome ,proteome as well as     metabolomic studies  are there.Further no studies exist as far as phageome[181] , fungal microbiome [182], as well as     archaeome [183] as well as     meta tanscriptome [184]. GM along with its products  remain to be a dark matter ,with our insight still restricted.

The other  restriction regarding,  GM composition  is that we don’t know regarding any nasal[185]   ,skin[186],or vaginal microbiota   studies [187] in this field of T1D  .These represent other significant  mucosal  surfaces   for   microbiota  -host Crosstalk    as well as     might participate   in the autoimmune  diseases like T1D.

It is significant  to realize that  no definite causal association isolated till now  with human disease along with microbiota.Rather than huge screening studies,a parallel strategy might be to concentrate on posit depending on particularmodes.The significance of microbiota  -host Crosstalk   in shaping the    GM composition,via both contact-based as well as     chemically modulated modes[188]. Current work   point that    community function ,instead of   particular taxonomic  composition might have an intrinsic part in disease  generation  [25].Overall ,future work needs to look into particular community Crosstalk   along with total function to yield better insight  of the differing  disease causations .Like it was recently posited that existent of microbes  that carry an insulin –like peptide  might  stimulate a ,molecular simulationmode in T1D as well as     found  viral insulitis[189].Further they revealed  the existence of  these viruses in human fecal as well as     plasma samples.

Further recently Kahalehili et al. posited that the immunoregulatory phytochemcial, indole-3-carbinol (I3C) which is existent  in cruciferous vegetables, would control the propagation of T1D in nonobese diabetic (NOD) mice. During digestion, I3C is metabolized into ligands for the aryl hydrocarbon receptor (AhR), a transcription factor that when systemically activated prevents T1D. In NOD mice, an I3C-supplemented diet led to strong AhR activation in the small intestine but minimal systemic AhR activity. In the absence of this systemic response, the dietary intervention led to exacerbated insulitis. Consistent with the compartmentalization of AhR activation, dietary I3C did not change T helper cell differentiation in the spleen or pancreatic draining lymph nodes.Rather , dietary I3C escalateded the percentage of CD4+RORγt+Foxp3- (Th17 cells) in the lamina propria, intraepithelial layer, and Peyer’s patches of the small intestine. The immune modulation in the gut was accompanied by alterations to the intestinal microbiome, with changes in bacterial communities seen  within one week of I3C supplementation. A transkingdom network was developed to anticipate  host-microbe interactions that were influenced by dietary I3C. Within the phylum Firmicutes, several genera (IntestinimonasRuminiclostridium 9, and unclassified Lachnospiraceae) were negatively regulated by I3C. Using AhR knockout mice, we validated that Intestinimonas is negatively regulated by AhR. I3C-mediated microbial dysbiosis was linked to increases in CD25high Th17 cells. Collectively, these data demonstrate that site of AhR activation and subsequent interactions with the host microbiome are important considerations in developing AhR-targeted interventions for T1D[190].

Moreover Calabrese et al reviewed the role of beneficial effects of Mediterranean diet along with prebiotics in prevention Type 1 diabetes mellitus (T1DM T1D) represents  a chronic autoimmune disease resulting from a complex interplay between genetic proneness along with environmental factors. Regarding the latter, gut microbiota has a key part  in the pathogenesis of T1DM, by affecting intestinal permeability, molecular mimicry, and modulating innate and adaptive immune system, as described in several previous studies. The composition of the gut microbiota is largely influenced by diet. Some observational studies have shown that a low fiber intake is associated with the development of many inflammatory   along with immune-modulateded diseases. In this regard , the Mediterranean diet (MD), which is based on high consumption of cereals (preferably as whole grains), legumes, nuts, vegetables, fruits, olive oil, and fish, could play a protective role. Many of the characteristic components of MD have functional characteristics with positive effects on health and well-being. Eating habits are the main significant determinants of the microbial multiplicity of the intestine and the food components influence both microbial populations and their metabolic activities from the early stages of life. Moreover, food metabolites influence the immune response. The intestine is considered the primary site where food metabolites mediate their effects, through epithelial integrity or mucosal immunity. The compromised epithelial integrity allows the translocation of bacteria and/or the diffusion of their products, such as food antigens and lipopolysaccharides, from the intestinal lumen to the tissues, which could enhance the stimulation of immune cells, contributing to the pathogenesis of autoimmune diseases, such as T1DM. The intake of a high amount of fiber and therefore of prebiotics with MD allows the microbiota to have a good microbial balance. Moreover, as more dietary fibers are ingested, a higher amount of short-chain fatty acids (SCFAs) is produced by anaerobic gut microbiota, promoting gut homeostasis, to which also contribute tryptophan metabolites and omega-3-fatty acids. Furthermore, the higher intake of polyunsaturated fatty acids and omega-3-fatty-acids contribute to a better metabolic control[191].

Again Verduci etal.emphasized on encouragement of long-term breast-feeding for at least the first 6 months of life and the prevention of early complementary foods along with  gluten introduction (before 4 months of age) as well as delaying cow milk introduction beyond  12 months of life. These detrimental feeding habits create a gut microbiota dysbiotic state that can contribute to the onset of T1D in infancy. Further , they  detailed  on the probability  of adding  probiotics, prebiotics and post-biotics in the avoidance of T1D especially in those high risk for T1D[192].

Giampioli tried to highlight the significance of FUT2 nonsecretory phenotype in high risk children. Children with non-secretor phenotype and those affected by T1D share low amounts  of bifidobacteria, along with lower of short-chain fatty acids amounts , intestinal phosphatase alkaline and a high incidence of inflammatory bowel diseases. In this  regard, host-gut microbiota dyad may represent a relevant contributor to T1D development along with propagation in view of  to its key part  in shaping host immunity and proneness to autoimmune conditions. The FUT2 gene is implicated for the composition and functional properties of glycans in mucosal tissues and bodily secretions, including human milk. FUT2 polymorphisms may considerably impact the  gut microbiota composition and host proneness to viral infections as well as  chronic inflammatory disease.They described how  probably crosstalk  among  mothers' phenotype, host FUT2 genetic background as well as   gut microbiota composition impact on T1D onset. The study of FUT2-gut microbiota interaction may add a new piece on the puzzling T1D etiology and unveil innovative targets of intervention to contrast T1D generation as well as  propagation. Dietary interventions, including the intake of α-(1, 2)-fucosyl oligosaccharides in formula milk and the use of specific prebiotics and probiotics, could be posited[193].

Further minimal knowledge   exists   in reference  to  the environmental factors that can be associated to these alleles.  Recent proof  pointed that, among those environmental factors, dysbiosis (imbalance) in the  gut microbiota might  participate  in the pathogenesis of T1D, affecting the integrity of the gut along with resulting in  systemic inflammation along with  auto-destruction of the pancreatic β cells. Various studies have  isolated alterations  in the gut microbiome composition in humans and animal models contrasingT1D subjects with controls. Those changes depicted by a greater  amounts  of Bacteroides  along with  lesser  amounts  of the butyrate-generating bacteria like  Clostridium clusters IV and XIVa.The modes by which the dysbiotic bacteria as well as  their metabolites crosstalk with the genome  and/or the epigenome of the host leading to destructive autoimmunity is still not clear. As T1Dis a multifactorial disease, understanding the interaction between different environmental factors  like  the gut microbiome, the genetic and the epigenetic determinants that are associated with the  early appearance of autoantibodies can expand our knowledge about the disease pathogenesis.Hence for better  understanding  into the crosstalk among  the gut microbiome, susceptibility genes,epigenetic factors, and the immune system in the pathogenesis of T1D  Elhag etal detailed the mode. It was also shown that the presence of a specific bacterial species such assingle-SFB) or Bacillus cereus in the female NOD mice procreated under germ-free conditions escalates the expression of the signature genes in Th17 cells   likeIl17a, Il17f, Il22, Il1r1, and Il23r which may delay the onset of T1D through regulating  the auto-immune response leading to enhanced gut integrity [reviewed in ref no 194 ]. Moreover, comparative   gut microbiota   evaluation  among the NOD mice with and without T1D revealed a   significantly greater  amounts  of four taxa that are associated with  antibiotic-mediated dysbiosis, including Enterococcus, Blautia, Enterobacteriaceae species, and A. mucinophila, these taxa  were shown to be associated with an accelerated progression into T1D (Figure 2) [reviewed in ref no 194 ].

An external file that holds a picture, illustration, etc.
Object name is ijms-22-00125-g002.jpg

Legend for Figure 2

Courtesy  ref no-194-Comparison between the opposite effects of probiotics and early-life use of antibiotics in NOD mice. (A) T1D protective effects of probiotic bacteria in NOD mice: Lactobacillus johnsonii N6 enhances the expression of INFγ and indolesamine 2,3-dioxygenase enzyme (IDO) which in turn increase the production of claudin-1 tight junction protein that maintains the integrity of the gut epithelium. Lactobacillus casei has probiotic properties, lowering the number of the splenic CD8+ cytotoxic T cells and promoting the expression of the anti-inflammatory cytokines (IL2, IL10) leading to increased immune tolerance in the gut. VSL3# probiotic has immune-modulatory functions, enhancing the expression of IL-33 cytokine that is necessary to maintain immune-tolerance in the gut and mesenchymal lymph nodes (MLN). (B) Role of antibiotic mediated dysbiosis in enhancing autoimmunity and pancreatic β-cells destruction in NOD mice: dysbiosis mediated by the use of antibiotics enhances the growth of pathogenic bacteria which promotes the inflammatory response via the TLR pathway which influences the gut permeability leading to metabolic endotoxemia. Endotoxemia stimulates the autoreactive T cells in the pancreatic lymph nodes leading to insulitis and β cell destruction.

Growing evidence indicates that these bacteria can modulate the gut inflammatory response,

either by increasing the gut permeability leading to metabolic endotoxemia orby enhancing the translocating of the LPS into the circulation which increases intestinalpermeability through a TLR-4 dependent activation of FAK-MyD88-IRAK4 signaling pathway,leading to the generation of insulitis and beta-cell destruction as shown in Figure2 [reviewed in ref no 194 ]. Moreever , peptidoglycan isolated from Firmicutes and Bacteroidetes species inAkita T1D mice were demonstrated to escalate  the risk of retinopathy in human retinal endothelial  cells [reviewed in ref no 194 ]. While the absence of the myeloid differentiation innate  immune adaptor gene (MyD88) in the NOD mice protects against T1D, it was observed   that this protection is mediated by the gut microbiome, through escalating the expression of the  immuno-regulatory enzyme indoleamine 2,3-dioxygenase (IDO) in the pancreatic lymphnodes [reviewed in ref no 194 ]. On the other hand, raising the MyD88-negative NOD mice under germfree  conditions quickly triggers the onset of T1D ,[reviewed in ref no 194 ]. Moreover, a recent  study revealed that raising of germ-free MyD88 knockout mice with a group of bacterial  probiotics or segmented filamentous bacteria provides either partial or complete protection  against T1D, highlighting the variability in the protection mechanism that is utilized by different   bacterial species [reviewed in ref no 194 ]. Progression into T1D in BDC2.5X TCR islet-specifictransgenic NOD mice raised under a specific-pathogen-free environment was shown to be  associated with an impaired gut barrier leading to microbiota-dependent endotoxemia, this endotoxemia can activate the migration of the islet autoreactive T cells to the pancreatic  tissue leading to the initiation of T1D [reviewed in ref no 194 ]. Additionally,   the protective  NOD mice  like  the Idd3/5 and C57BL/6 mice, (which carry T1D protective alleles) have   a distinct gut microbiome composition when compared to wild type NOD mice, showing  differences in the relative abundance of Lactobacillus, S24-7, and Ruminococcus ..Although there are major variations amongst human and animal models in T1D  correlated gut microbiota, some commonalities emerge mainly in the mechanism by which   different microbiomes can influence the controlling functions of the immune system, mainly  through the role of LPS and viral proteins which are known to be associated with the   initiation of T1D [reviewed in ref no 194].

Thus finally concluding ,still lot of work needs to be done with regards to correlation of susceptibility genes,environmental factors along with solving the mystery of gut microbiota in initiation of T1D.Yet ensuring breast feeding trying pre/probiotics and diet changes like MD ,or FUT2 / AhR activation   targeting might aid in helping till further insight is gained.

References

  1. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. (2017). An Update on Etiopathogenesis and Management  of Type 1 Diabetes Mellitus; Open Access Journal of Endocrinology, 1(2):1-23.
  2. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. (2019). Restricting Carbohydrates in the Diet-A Possible Method of Meeting the  Challenges of Increasing Diabesity in Type1 Diabetes along with  Meeting Exercise Performances Requirements-A Review. J Endocrinol,  3(1): 000138.
  3. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. (2020). ’How can we use Empagliflozin as an adjuvant in reducing required need of insulin in type1 diabetes along with lowered HbA1c,weight without fear of DKA.-A Minireview’’. J Clin Cases Rep, 4(2): 30-38.    
  4. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. (2020). An update on the Immunotherapy Strategies for the treatment of Type 1 Diabetes (TID)-How far have we reached in reaching insulin independency in TID therapy-A Systematic Review. J Endocrinol, 4(1): 000149.
  5. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. Attempting Getting Insulin Independent Immunotherapies in Type 1 Diabetes Mellitus (T1D) in the Pre Stage 1 (BeforeIslet Autoantibodies)”. Acta Scientific Paediatrics 2020;3(6): 01-04.
  6. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. Path Directed towards a Stage when we almost Cure Type1 Diabetes Mellitus (T1dm) after a Century of Insulin Advent”. EC Diabetes and Metabolic Research 2020;4(6): 37-46.
  7. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. Utilization of Extracellular Vesicles for Treatment of Type 1 Diabetes Mellitus (T1DM) Along with Type 2 Diabetes Mellitus (T2DM) besides Complications Associated with Diabetes- A Systematic Review. J Clin DiabetesObes.2020.1.001-013. DOI: 10.47755/
  8. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. (2020). The association of dietary fatty acids and gut microbiota alterations in the development of neuropsychiatric diseases: A systematic review. Obes Res Open J. 7(1): 19-45.
  9. Kulvinder Kochar Kaur,Allahbadia GN,Singh M.. Have Probiotics and Synbiotics passed the test of time to be implemented in management of obesity and related metabolic disorders-a comprehensive review. Adv Obes Weight Manag Control. 2019;9(1):21‒28. DOI: 10.15406/aowmc.2019.09.00269.
  10. Kulvinder Kochar Kaur,Allahbadia GN,Singh M. Will Probiotics Provide the Answer for Therapy of Non-alcoholic Fatty Liver Disease (NAFLD)? – A Systematic Review. Biochem Physiol  2020;9: 257.
  11. Kulvinder Kochar Kaur,Allahbadia GN,Singh M.’Advances in Probiotics use  with the utilization of engineering technology for diseases beyond Obesity ,Non Alcoholic Fatty Liver Disease ,  to treat neurodegenerative diseases,metabolic diseases like Type1 diabetes ,infectious diseases and infections –A Systematic Review’’. J Endocrinol 2020;4(1):1-12.
  12. Kulvinder Kochar Kaur,Allahbadia GN,Singh M.Weight loss Associated with high protein Intake in Obesity:Interactions of Gut Microbiota in Protein Sources  influencing this positive effect. Acta Scientific Nutritional Health 2018;2(7) : 80-89
  13. Blaum HE.The human microbiome  Adv Med Sci 2017;62:414-20.
  14. Gordo I.Evolutionaru change in the human  gut microbiome:  From a static to a dynamic view.PLoS Biol 2019;17:e3000126.
  15. De Luca F,Shofield Y.The Microbiome in autoimmune diseases.Clin Exp Immunol   2019;194:74-85.
  16. Maruvada P,Leone V,Kaplan LM,,Chang EB.The human microbiome  and obesity:moving beyong associations  .Cell Host Microbe  2017;22:589-99.
  17. Rinaldi M,Perricone R,Blank M, Perricone  C, Shofield Y.Anti Saccharomyces  cerevistae  auto- antibodies in autoimmune diseases:from bread baking to  Autoimmunity. ClinRev A llergy   Immunol   2013;45:152-61.
  18. Gevers D,Kugathasan S,Denson LA,Vasquez –Baeza Y,Van Treuren W,Rden B,etal.The treatment –naive microbiome  in new onset Crohn’s disease . Cell Host Microbe  2014;15:382-92.
  19. Carding S,Verbeke K,Vipond DT,Corfe BM,Owen LJ.Dysbiosis  of the gut microbiome in disease. Microb Ecol Health Dis 2015;26:26191.
  20. Navaneethan U,Venkatesh PG,Shen B. Clostridium difficile infection and inflammatory bowel disease:understanding the evolving relationship.World J Gastroenterol  2010;16: 4892-904.
  21. Quagliarello A,Aloisio I,Bozzi Cionci N,Luiselli D,D’auria G,Martinez Priego L,etal.Effect of Bifidobacterium breve  on the intestinal  Microbiata of celiac  children on a gluten free diet:a pilot study .Nutrients 2016;8: 660.
  22. Heinz-BuschartA,May P,Lazny CC,Lebrun LA,Bellora C,Krishna A,etal.Erratum:integrated multi-omics of the human gut microbiome in a case study of familial Type 1 diabetes. Nat Microbiol  2016;2: 16227.
  23. Vatanen T,Franzosa EA,,Schwager R,Tripathi S,Arthur TD,Vehik K,etal. The human  gut microbiomein early onset Type 1 diabetes mellitus from the TEDDY study.Nature 2018;562:589-94.
  24. Atarashi K,,Honda K. Microbiata in Autoimmunity and tolerance Curr Opin Immunol   2011;23:761-8.
  25. RussellJT,Roesch LFW,Ordberg M,Ilonen J,Atkinson MA,Schatz DA,etal.Genetic risk for Autoimmunity is associated  with distinct changes in human  gut microbiome.Nat Commun 2019;10:3621.
  26. Wu HJ,Ivanov LI,Darce I,Hattori K,Shima T,Umesaki Y,etal.Gut-residing segmentous filamentous bacteria drive autoimmune arthritis via Thelper 17 cells . Immunity 2010;32: 815-27.
  27. Atarashi K,Tanoue T, Shima T,Imaoka A,Kuwahara T,Momose Y,etal.Induction of colonic regulatory Tcells by by indigenous Clostridium species.Science 2010;331: 337-41.
  28. Ceccarelli F,Agmon-Levin N, Perricone  C. Genetic  factors of autoimmune diseases.J ImmunolRes   2017;2017:2789242.
  29. Jorg S,Grohme DA,Erzler M,Binsfeld M,Haghikia A,Muller DN,etal. environmental factors   in autoimmune diseases and their role in Multiple Sclerosis.Cell Mol Life Sci 2016;73: 4611-22.
  30. Gets DR,Chastain EM,Terry RL,Miller SD.Virus infection,anti Viral immunity and autoimmunity. Immunol    Rev 2013;255: 197-209.
  31. Vanderlukt CJ, Miller SD.Epitope spreading . Curr Opin Immunol   1996;8:831-6.
  32. De Goffau MC,Luopajarvi K,Knip M, Ilonen J,Ruohtula T,Pesola I, VirtanenSM,Harkonen T,etal.Fecal Microbiota composition   differs between children with β-cell auto immunity and those without .  Diabetes 2013;62:1238-44.  
  33. Belkaid Y,Hand TW.Role of Microbiata in immunity and inflammation.Cell 2014;157: 121-41.
  34. De Goffau MC,Fuentes S,Van Den Bogert B,Honkanen H,De Vos WM.Welling GW,etal.Aberrant gut microbiota  at the onset of Type 1 diabetes in young children .Diabetologia 2014;57: 1569-77.
  35. De Groot  PE,Belzer C,Aydin O,Levin E,Levels JH,Aalvink S,etal.Distinct faecal and oral Microbiata composition in human Type 1 diabetes,an observational study. PLoS One  2017;12:e0188475.
  36. Maahs DM,West NA,Lawrence JM,Mayer-Davis EJ.Epidemiology of  type 1 diabetes  . Endocrinol Metab  Clin N Am2010;39:481-97.
  37. Patterson CC,Dahlquist GG,Guyrus E,Green A,Soltesz G,Group ES.Incidence,trends for childhood Type 1 diabetes in Europe during 1989-2003 and predicted new cases 2005-20:A multicenter prospective registration study.Lancet 2009;373: 2027-33.
  38. Jin JS,Touyama M,Yamada S,Yamazaki T,Benno Y.Alteration of a human intestinal   Microbiata  under extreme life environment in the Antarctica . Biol Pharm  Bull 2014;37:1899-906.
  39. Soderstrom U,Aman I,Hjern A,.Being born in Sweden increases the risk of Type 1 diabetes-a study of migration of children to Sweden as a natural experiment.Acta Paediatr 2012;101:73-7.
  40. Voight RM,Forsyth CM, Green SJ,Mutlu E,Engen P,Vitaterna MH,etal.Circadian disorganization of intestinal   Microbiata  . PLoS One  2014;9:e97500.
  41. Voight RM,Forsyth CM, Green SJ,Mutlu E,Engen P,Keshvarzian A. Circadian rhythm and the gut microbiome. Int  Rev Neurobiol 2016;131:193-205.
  42. Marre ML,James EA,Piganelli JD.Beta cell ERStress and the Implications for immunogenicityin Type 1 diabetes.Front Cell Dev Biol  2015;3:67
  43. Atkinson MA,Gale EA.Infant dies and Type 1 diabetes:too early .too late or just too complicated ?JAMA 2003;290: 1771-2.
  44. Purcell AW,Sechi S,Dilorenzo TP.The evolving landscape  of autoantigens discovery  and characteristization of Type 1 diabetes. Diabetes 2019;68:879-86.
  45. Delong T,Wiles TA,Baker RL, Bradley B,Barbour G,Reisdorph R,etal.Pathogenic  CD4+T cells in Type 1 diabetes recognizes epitopes  formed by peptide fusion. Science 2016;351: 711-4.
  46. GroupSFDIYS ,Liese AD,D’agostino RB Jr,Hamman RF,Kilgo PD,Lawrence JM,etal. The burden of diabetes mellitus among US youth :prevalence  estimates from the search for Diabetes in youthstudy,Paediatrics 2006;118: 1510-8.
  47. Bingley PJ, Gale EA.Rising incidence of IDDM  in Europe . Diabetes Care  1989;12:289-95.
  48. Gale EA.The rise of childhood Type 1 diabetes in the 20th century. Diabetes 2002;51:3353-61.
  49. LudwigssonJ.Increasing incidence but decreasing awareness  of   Type 1 diabetes in Sweden. Diabetes Care  2017;40:e143-4.
  50. Diabelea D.The accelerating epidemic of  childhood Diabetes.Lancet 2009;373: 1999-2000.
  51. Redondo MJ,Jeffrey J,Fain PR, Eisen barth GS.,Orban T,Concordance for islet autoimmunity among monozygotic twins. N Engl J Med 2008;359:2849-50.
  52. Thayer TC,Wilson SB,Mathews CE.Use of in non obese diabetic  mice to understand human type 1 diabetes. Endocrinol Metab Clin North  Am   2010;39:541-61.
  53. Wicker LS,Clark J,Fraser HI,Garner VE,Gonzalez—Munoz A,Healy B,etal. Type 1 diabetes genes  and pathways shared  by humans and NOD mice .J Autoimmun 2005;25(Suppl):29-33.
  54. Pozzill P,Signore A,Williams AJ,Beales PE. NOD mouse   colonies around the world –recent facts and figures. Immunol Today    1993;14:193-6.
  55. Wen L,Ley RE,Volchkov PY,Stranges PY,Avanesyan L,Stonebraker AC,etal. Innate    immunity and intestinal  Microbiata in the development of Type 1 diabetes. Nature 2008;455:1109-13.
  56. Markle JG,Frank DN,Mortin –Toth S,Robertson CE,Feazel LM,Rolle-Kampczyk U,etal.Sex differences in the Gut Microbiome   drive hormone Dependent   regulation of autoimmunity. Science 2013;339: 1084-8.
  57. Onengut –Gumuscu S,Chen WM,Burren O,Cooper NJ,,Quinlan AR,Mychalecky JC,etal.Fine mapping of Type 1 diabetes susceptibility loci  and evidence for colocalization of causal variants  with lymphoid  gene enhancers.Nat Genet 2015;47: 381-6.
  58. Hagopian WA,Erlich H,Lenmark A,Rewers M,Ziegler AG.,Simell O,etal. The environmental determinants of  diabetes in the Young(TEDDY):Genetic criteria  and international diabetes risk screening  of 421,000 infants. Paediatr Diabetes   2011;12:e733-43.
  59. Rewers M , LudwigssonJ. Environmental  risk  factors for   Type 1 diabetes. Lancet 2016;387: 2340-8.
  60. Stewart CJ,Ajami NJ,O’Brien JL,Hutchinson DS,Smith DP,Wong MC,etal.Temporal development of the Gut Microbiome    in early childhood from the TEDDY  study . Nature 2018;562:583-8.
  61. Kondrashova A,Reunanen A,Romanov A,Karvonen A,Viskari H,Vesikari T,etal.A six-fold gradient in the incidence of Type 1 diabetes at the eastern  border of Finland.Ann Med 2005;37:67-72.
  62. Gale EA.A missing link in Hygiene hypothesis? Diabetologia 2002;45:588-94.
  63. Bach JF.Six questions about the Hygiene hypothesis? CellImmunol 2005;233:158-61.
  64. D’Angeli MA,Merzon E,Valbuena LE,Tirschwell D,Paris CA,Mueller BA. Environmental   factors  associated  with   childhood onset   Type 1 diabetes:an exploration of the  Hygiene  and overload hypothesis .Arch  Paediatr Adolesc Med 2010;164:732-8.
  65. Briton RA,Young VB.Role of intestinal  Microbiata in resistance to colonization  by Clostridium difficile. Gastroenterology   2014;146: 1547-53.
  66. Kostic AD,Gevers D,Silzander H, Vatanen T,Hyotylainen T,Hamalainen AM,etal. The dynamics of the human infant  Gut Microbiome     in  development  and early progression of Type 1 diabetes. Cell Host Microbe  2015;17:260-73.
  67. Dedrick S,Sundaresh B,Huang Q,Brady C,Yoo T,Cronin C,etal.The role of Gut Microbiota and environmental factors   in Type 1 diabetes  pathogenesis.Frontiers Endocrinol 2020;11:78.
  68. Vatanen T , Kostic AD,D;Hennezel E, Silzander H, Franzosa EA,Yassour M,etal.Variations in Microbiome LPS immunogenicity  contributes to autoimmunity in humans . Cell   2016;165:842-53.
  69. Akerman L, LudwigssonJ,Swartling U,Casas R. Characteristics of the pre diabetic period in children with high risk of   Type 1 diabetes recruited  from the general Swedish population-The ABIS Study. Diabetes Metab Res Rev  2017;33.
  70. KubinakJL,Stephens WZ,Soto R,Petersen C,Chiaro T,Gogokhia L ,etal.MHC Variation sculpts individualized  microbial communities  that control susceptibility to enteric infection . Nat Commun 2015;6:8642.
  71. Farsejo AO, LudwigssonJ.Pet exposure in the family during pregnancy  and risk for   Type 1 diabetes-The prospective ABIS Study. Paediatr Diabetes   2018;19:1206-10.
  72. Tun HM,Konya T,Takaro TK,Brook JR, Chari R,Field CJ,etal.Exposure to household furry pets influences the Gut Microbiota of infants at3-4mths following various birth scenarios. Microbiome 2017;5:40.
  73. Walker WA.The importance of appropriate initial colonization  of the intestine in newborn,child ,and adult health Paediatr Res  2017;82: 387-95
  74. Blasser MJ,Dominguez-Bello MG.The human Microbiome before birth . Cell Host Microbe  2016;20:558-60.
  75. Koren O,Goodrich JK,Cullender TC,Spor A,Laitinen K,Backhead HK,etal.Host remodeling of Gut Microbiome   and metabolic changes  during pregnancy . Cell   2012;150:470-80.
  76. Gronlund MM,Lehtonen OP,Eorela E,Kero P.Fecal Microbiota in healthy infants born by different methods of delivery:permanent changes in intestinal flora after caesarean delivery .J Paediatr Gastroenterol Nutr    1999;28: 19-45.
  77. Dominguez-Bello MG,Costello EK,Contreras M,Magris M,Hidalgo G,Fierer N,etal.Delivery mode shapes the initiation and structure of theinitial   Microbiata across multiple habitats in the newborn. Proc NatlAcad Sci USA 2010;107:11971-5.
  78. Kuhle S,Tong OS,Woolcott CG.Association between caesarean section and childhood obesity:a systematic review and meta-analysis.Obes Rev 2015;16: 295-303.
  79. Tun HM,Bridgman SL, Chari R,Field CJ,Guttman DS,Becker AB, etal.Roles of birth mode and infant   gut Microbiata in intergenerational transmission of overweight and obesity from mother to offspring .JAMA Paediatr2018;172: 368-72.
  80. Adlerberth I,Lindberg E,Aberg N,Hesselmar B,Saalman R,Strannegard IL, etal.Reduced enterobacterial and increased staphylococcal colonization of  o the infant bowel:an effect of hygienic lifestyle? Paediatr  Res 2006;59: 96-101.
  81. RutaysireE,Huang K,Liu Y,Tao F.The mode of delivery and colonization of   the gut Microbiata during the first year of infant’slife:a systematic review .BMC Gastroenterol 2016;16: 86.
  82. Dogra S,Sakwinska O,Soh SE,Ngom Bru C,Bruck WM,Berger B, etal.Dynamics of infant   gut Microbiata are influenced by Delivery mode and gestational duration  and are  associated with subsequent adiposity.M Bio2015;6: e02419-14.
  83. Cardwell CR,Stene LC,Joner G,Cinek O,Svensson J,Goldacre MJ, etal. Caesarean section  is associated with an increased risk of childhood onset- Type 1 diabetes: meta-analysis  of observational studies. Diabetologia 2008;51:726-35.
  84. Magne F,Puchi Silva A,Carvachal B,Gotteland M.The elevated rate of Caesarean section  and its contribution to non-communicable chronic diseases in Latin America :the growing involvement of the Microbiata.Front Paediatr  2017;5: 192.
  85. Samuelsson U,Lindell N,Bladh M,Akesson K,Carlsson A,Josefsson A. Caesarean section   per se does not  increase  the risk of  offspring developing  Type 1 diabetes:a Swedish  population based study . Diabetologia 2015;58:2517-24.
  86. Hansen CH,Andersen LS,Krych L,Metzdorff SB,Hasselby JP,Skov S , etal. Mode  of  Delivery  shapes gut  colonization  pattern and modulates  regulatory immunity in mice . J Immunol 2014;193:1213-22.
  87. He B,Xu W,Santini PA,Polydorides AD,Chiu A,Estrella J, etal. Intestinal  bacteria triggers  T –cell independent immunoglobulinA2  class switching by inducing epithelial –cell secretion of the cytokine  APRIL .Immunity 2007;26:812- 26.
  88. Lutgendroff F, Akerman LM,Soderholm JD.The role of Microbiota and probiotics in stress induced gastrointestinal damage. Curr Mol Med 2008;8:282-98.
  89. Mazmanian SK,Round JL,Kasper DL.A microbial symbiosis factor prevents intestinal  inflammatory disease. Nature 2008;453:620-5.
  90. Nejentsev S,Sjoroos M,Soukka  T,Knip M,Simell O,Lovgren T, etal.Population based genetic screening  for the estimation of   type 1 diabetesmellitus risk in Finland :selective genotyping in markers of the HLA-DQB1, HLA-DQA1,and HLA-DRB1loci. DiabetMed  1999;16:985-92.  
  91. Giongo A,Gano KA,Crabb DB,Mukherjee N, Novelo LL,Casella G etal.Toward defining the autoimmune microbiome  for   type 1 diabetes.ISME J 2011;5:82-91.
  92. Davis –Richardson AG,Aedissone AN,Dias R, Simell V,Leonard MT,Kempainnen KM, etal. Bacteroidesdorei dominates Gut Microbiome   prior to auto immunityin Finnish children at high risk for type 1 diabetes. Front Microbiol   2014;5: 678.
  93. Stene LC,Magnus P,Lie RT,Sovik O,Joner G.Norwegian childhood Diabetes Study G.No association between preeclampsia or Caesarean section    and incidence of type 1 diabetes among children:a large population based cohort study . Paediatr  Res 2003;54:487-90.
  94. Knol J,Scholtens P,Kafka C,Steenbakkers J,Gro S,Helm K, etal.Colon microflora in infants fed formula  woth galactoand fructo-oligosaccharides:more like breast fed infants more like breast fed infants.J Paediatr Gastroenterol  Nutr 2005;40: 36-47.
  95. Palmer C,Bik EM,Diguilo DM,Relman DA,Brown PO.Development of the human infant intestinal  Microbiota. PLoS Biol 2007;5:e177.
  96. LopezNM,Ponce S,Piccininini D,Perez E,Roberts N.From hospital to home care:creating a domesticenvironment for elderly and disabled people IEEE Pulse  . 2016;7: 38-41.
  97. Pannaraj PS,Li F,Cerini C,Bender JM,Yang S,Rollie A, etal. Association  between breast milk communities and establishment and development of the infant Gut Microbiome  . JAMA Paediatr2017;171: 647-54.
  98. Solis G,DeLos Reyes-Gavilian CG,Fernandez N,Margolles A,Gueimonde M.Establishment  and development  of lactic acid bacteria and BifidobacteriaMicrobiota in breast milk and infant gut.Anaerobe  2010;16: 307-10.
  99. .Gibson GR,Wang X.Regulatory effects of Bifidobacteria on the growth of colonic bacteria.J Appl Bacteriol 1994;77: 412-20.
  100. Fujiwara S,Hashiba H,Hirota T,Forstner JF.Proteonaceous factor(s)in culture supernatant  fluids of Bifidobacteria which prevents the binding of  enterotoxigenic Escherichia Coli togangliotetraosylceramide . Appl   EnvironBacteriol 1997;63: 506-12.
  101. Leivin V,Peiffer I,Hudault S,Rochat F,Brassart D,Nesser JR, etal. Bifidobacterium strains from resident  human gastrointestinal microflora exert antimicrobial activity .Gut 2000;47: 646-52.
  102. Picard C,Fioramonti J,Francois A,Robinson T,Neant F,Matuchansky C.Review article: Bifidobacteria as probiotic agents- physiologicaleffects and clinical benefits.Aliment  PharmacolTher  2005;22:495-512.
  103. Insel R,Knip M.;Prospects for primary prevention of Type1 Diabetes by restoring a disappearing microbe. Paediatr Diabetes   2018;19:1400-6.
  104. Hooper LV,Littman DR,Macpherson AJ.Interactions between the Microbiota and the immune system. Science 2012;338: 1268-73.
  105. Alves JG,Figuereiroa JN,Meneses J, Alves GV. Breast feeding     protects against type1 diabetes  mellitus:a case-sibling study . Breastfeed  Med 2012;7: 25-8.
  106. AzadMR,Konya T,Maughan H,Guttman DS,Field CJ,Chari RS, etal. Gut Microbiotaof healthy Canadian infants:profile by Mode  of  Delivery and infants diet at 4 mths .CMAJ 2013;185: 385-94.
  107. Albenberg LG,WuGD.Diet and intestinal  Microbiome: associations ,functions and implications for health and disease. Gastroenterology  2014;146:1564-70.
  108. Madan JC,Hoen AG,Lundgren SN,Farzan SE,Cottingham KL,Morrison HG, etal. Associationof  Caesarean Delivery   and formula supplement with the intestinal  Microbiome  of 6weeks infants. JAMA Paediatr2016;170: 212-9.
  109. Lund-Blix NA,Stene LC,Rasmussen T,Torjesen PA,Andersen LE,Ronningen KS. Infant feeding in relation toislet autoimmunity  and Type1 Diabetes in genetically susceptible  children ;the MIDIA Study. Diabetes Care  2015;38:257-63.
  110. Wahlberg J, Vaarala O LudwigssonJ, GroupAB-S.Dietary risk factors for the emergence of Type1 Diabetes –related antibodies . Br J Nutr 2006;95: 63-8.
  111. Holmberg H,Wahlberg J, Vaarala O LudwigssonJ ,GroupAB-S.Short duration of Breast feeding      as a risk factor for  beta cell- autoantibodies  in 5 year old children from the general population.Br J Nutr 2007;97: 111-6.
  112. Vatanen T , Kostic AD,D;Hennezel E, Silzander H, Franzosa EA,Yassour M,etal.Variation in microbiome LPS immunogenicity contributes  to autoimmunity in humans.Cell   2016;165: 1551.
  113. Meija –Leon ME,Petrosino JF,Ajami NJ,Dominguez-Bello MG,De la Barca AM.Fecal Microbiotaimbalance in Mexican children  with Type1 Diabetes.Sci Rep   2014;4: 3814.
  114. Lu YC,Yeh WC,Ohashi PS.LPS/TLR4 signal transduction pathway.Cytokine  2008;42: 145-51.
  115. Sai P,Rivereau AS.Prevention of Diabetes in the non obese diabetic mouse  by oral immunological treatment.Comparative efficiency of human insulin and two bacterial antigens, lipopolysaccharide from Escherichia Coli and glycoprotein extract from Klebsiella pneumonia. Diabetes Metab 1996;22:341-8.
  116. Guilden E,Ihira  M,Ohashi A,Reinbeck AL,Freudenberg MA,Kolb H,etal. Toll like receptor-4 deficiency accelerates the development of insulin-deficient Diabetes in  non obese diabetic  mice . PLoS ONE  2013;8:e75385.
  117. Frese SA,Hutton AA,Contreras LN,Shaw CA,Palumbo MC,Casaburi G,etal.Persistence of supplemented Bifidobacterium longumsubsp .infantis EVC001 in breast fed infants.mSphere  2017;2: e00501-17.
  118. VirtanenSM,Knip M.Nutritional risk predictors of  beta cell auto immunity and  Type1 Diabetes at a young age.Am J Clin Nutr2003;78:1053-67.
  119. Knip M, VirtanenSM,Seppa K, Ilonen J,Savilahti E, Vaarala O, etal. Dietary  intervention  in infancy and later signs of  beta cell auto immunity . N Engl J Med 2010;363:1900-8.
  120. Kolb H,Burkart V.Nicotinamide in-  Type1 Diabetes.Mechanism of action revisited  . Diabetes Care 1999;22(Suppl 2):B16-20.
  121. Ho E,Quan N,Tsai YH,Lai W,Bray TM. Dietary   zinc supplementation  minhibits NF kappa B activation  and protects against chemically induced Diabetes  in CD1 mice . Exp  Biol  Med 2001;226:103-11.
  122. Gale EA ,Bingley PJ,Emmett CL,Collier T.European Nicotinamide Diabetes Intervention TrialG. European Nicotinamide Diabetes Intervention Trial(ENDIT):A randomized controlled trial of   Intervention   before the onset of Type1 Diabetes.  Lancet 2004;363: 925-31.
  123. Luong K,Nguyen LT, Nguyen DN.The role of  vitamin D  in protecting Type1 Diabetes. Diabetes Metab   Res Rev 2005;21:338-46.
  124. Zipita CS,Akobeng AK. Vitamin D supplementation in early childhood  and risk of   Type1 Diabetes :A systematic review and meta-analysis.Arch Dis Child2008;93:512-7.
  125. Cantorna MT. vitamin D   and autoimmunity :is Vitamin D  status an environmental factors   affecting autoimmune  diseases  prevalence.Proc Soc Exp Biol MED 2000;223:230-3.
  126. Scott FW.Cow milk and insulin dependent diabetes:is there a relationship. AmJ Clin  Nutr 1990;51: 489-91.
  127. Murri M,.Leiva I,Gomez-Zumaquero JM,Tinahones FJ,Cardona P,Sonquer F, etal. Gut Microbiota in children with Type1 Diabetes differs from  that in healthy  children:acase control study.BMC Med  2010;363:589-98.
  128. Fasano A.Zonulin  and its regulation of intestinal barrier function:the biological door  to inflammation, autoimmunity and cancer.Physiol Rev 2011;91:151-7.
  129. Marietta EV,Gomez AM,Yeoman C,Tilahun AY,Clark CR,Luckey DH, etal. Low incidence of spontaneous   Type1 Diabetes  in non obese diabetic   mice raised on Gluten  free  diets is associated   with  changes in the intestinal microbiome . PLoS ONE  2013;8:e78687.
  130. Qin H,Zhang Z,Hang X,Jiang Y.L.plantarium prevents enteroinvasive  Escherichia Coli induced  tight junction proteins changes in intestinal epithelial cells . BMC Microbiol  2009;9:63.
  131. Pastore MR,Bazzigalappi E,Belloni C,Arcovio C,Bonifacio E,Bosi E.Six months of Gluten  free  diet do not influence   autoantibody  titres, but improve insulin secretion in subjects at high risk  for  Type1 Diabetes . J Clin Endocrinol Metab  2003;88:162-5.
  132. Saadah OI,Zacharin M,O’Callaghan A,Oliver MR,,Catto-Smith AG.Effect of Gluten  free  diet and adherence to growth  and diabetic control in diabetics with celiac disease. Arch Dis Child2004;89:871-6.
  133. Rami B,Sumnik Z,Schober E,Waldhor T,Battelino T,Bratanie T, etal. Screening detected celiac disease in  children with Type1 Diabetes mellitus :effect on the clinical course in (a case control study). J Paediatr Gastroenterol  Nutr 2005;401: 317-21.
  134. Ziegler AG,Schmid S,Huber D, Bonifacio E . Early  infant feeding and risk of developing  Type1 Diabetes –associated autoantibodies .JAMA   2003;290:1721-8.
  135. Schmid S,Koczwara K,Schwinghammer S,Lampasona V, Ziegler AG, Bonifacio E .Delayed exposure  to wheat and barley proteins  reduces  Diabetes  incidence  in non obese diabetic   mice. Clin Immunol 2004;111:108-18.
  136. Mueller DB, Koczwara K, Mueller  AS,Pallauf I, Ziegler AG, Bonifacio E .Influence of Early  nutritional  components on the development  of murine   auto immune Diabetes.Ann Nutr Metab  2009;54:208-17.
  137. Antvorskov JC,Josefsen K,HauptJorgensen M,Fundova P,Funda DP,Buschard K. Gluten  free   diet only during pregnancy  efficiently prevents Diabetes in NOD mouse offspring.J   Diabetes  Res 2016;2016:3047574.
  138. Antvorskov JC,Halldorsson TI,Josefsen K,Svensson J,Granstrom C,Roep B, etal. Association between  maternal   Gluten intake and  Type1 Diabetes  in offspring :national  prospective  cohort study in Denmark.BMJ  2018;362:K3547.
  139. VirtanenSM,Uusitalo L,Kenward MG,Nevalainen J, UusitaloU,Kronberg-Kippilla C, etal. Maternal food consumption during pregnancy and risk of advanced beta cell autoimmunity in the offspring  . Paediatr Diabetes   2011;12:95-99.
  140. Svensson J ,Sildorf SM,Pipper CB,Kyvsgaard JN,Bojstrup J,Pciot FM, etal. Potential beneficial  effects of a gluten free diet  in newly diagnosed  children with Type1 Diabetes  a pilot study.Springer Plus   2016;5:994.
  141. Paun A,Yau C,Danska JS.The influence of the Microbiome  on Type1 Diabetes  .J Immunol 2017;198:590-5.
  142. Montoni S,Cocco P,Aru G,Cucca F. Nutritional  factors and worldwide incidence of childhood  Type1 Diabetes  . AmJ Clin  Nutr 2000;71: 1525-9.
  143. Elliot RB,Harris DP,Hill JP,Bibby NJ,Wasmuth HE.Type 1 (insulin dependent) diabetes mellitus and  cow’s milk :casein variant composition . Diabetologia   1999;42:292-6.
  144. VirtanenSM, Nevalainen J, Kronberg-Kippilla C, Ahonen S,Tapanainen H,Uusitalo L, etal.Food consumption and advanced beta cell autoimmunity in children with HLA P-conferred susceptibility to Type1 Diabetes  :a nested case control design. AmJ Clin  Nutr 2012;95: 471-8.
  145. Savilahti E,Saukkonen TT,Virtala ET,Tuomilehto J,Akerblom HK.Increased  levels of cow’s milk and beta lactoglobulin antibodies in young  children with newly diagnosed IDDM.TheChildhood  Diabetes in Finland Study Group. Diabetes   Care 1993;16:984-9,
  146. Robertson L,Harrild K.Maternal and neonatal risk factors  for Childhood Type1 Diabetes  :a matched case control study.BMC Public Health 2010;10: 281.
  147. Savilahti E,Saa rinen KM.Early infant feeding  and Type1 Diabetes  . Eur J   Nutr 2009;48: 243-9.
  148. Writing Group for the TSG, Knip M, Akerblom HK,Al Taji E,Becker D,Bruining J etal.Effect of hydrolyzed    infant  formula   vs conventional  formula on risk of Type1 Diabetes  :the TRIGR randomized clinical  trial .JAMA  2018;319:38-48.
  149. Vaarala O, Ilonen J,Ruohtula T,Pesola I, VirtanenSM,Harkonen T,etal.Removal of bovine insulin  from cow’s milk formula and early initiation of beta cell  autoimmunity   in the FINDIA pilot study . Arch  Paediatr Adolesc Med 2012;166:608-14.
  150.  
  151. Palleja A,Mikkelson KH,Forslund SK,Kashani A,Allin KH,Nielsen T etal.Recovery of Gut Microbiota of healthy adults  following antibiotic exposure.Nat Microbiol 2018;3:1255-65.
  152. Hicks LA,Taylor THJr,Hunkler RJ.US outpatient  antibiotic prescribing ,2010. N Engl J Med 2013;368:1461-2.
  153. Bokulich NA,Chung J,Battaglia T,Henderson T,Jay M,Li H etal. Antibiotics ,birth mode ,and diet shape microbiome maturation  during Early life . Sci Transl Med 2016;8:342ra382.
  154. Tapia G,Stordal K,Marild K,Kahrs CR,Srivarhaug T,Njolstad PR, etal. Antibiotics ,Acetaminophen and infections during prenatal and early life in relation to Type1 Diabetes  .Int J Epidemiol  2018;47:1538-42.
  155. Kempainnen KM,Vehik K,Lynch KE,Larsson HE,Canepa R,Smell VE, etal. Association between early life  Antibiotics use  and the risk of    islet or Coeliac disease Autoimmunity.JAMA Paediatr2017;171:1217-25.
  156. Mikkelsen KH,KnopFK,Vilsboll T,Frost M,Hallas J,Pottegard A.Use of    Antibiotics in Childhood and risk of Type1 Diabetes:a  population based case  control  study. Diabet Med 2017;34:272-7.
  157. Kilkkinen A, VirtanenSM, Kenward MG,Salkinoja-Salonen M,Gissler M, etal.Use of anti Microbials and risk of Type1 Diabetes in a population based mother-child cohort. Diabetologia 2006;49:66-70.
  158. Boursi B,Mamtani R,Haynes K,Yang YX.The effect of past Antibiotics exposure on Diabetes risk.Eur J Endocrinol 2015;172:639-48.
  159. Brugman S,Klatter EA,Visser JT,Wildeboer –Veloo AC,Harmsen AJ,Rozing J, etal. Antibiotic treatment partially protects against Type1 Diabetes   in the Bio-breeding  Diabetes prone rats .Is the gut flora involved in the  development of Type1 Diabetes   ? Diabetologia 2006;49:2105-8.
  160. Hara N,Alkanani AK,IrD, Robertson CE,Wagner BD,Frank DN, etal.Prevention of virus induced  Type1 Diabetes with Antibiotics therapy .J Immunol  2012;189:3805-14.
  161. Livanos AE,Greiner TU,Vangay P,Pathmasiri W,Stewart D,Mcritichie S, etal. Antibiotic mediated Gut Microbiome perturbations  accelerates development of Type1 Diabetes inmice. Nat Microbiol 2016;1:16140.
  162. Hansen CH,Krych L,Nielsen DS,Vogensen FK, HansenLH,Sorensen SJ, etal. Early life treatment with vancomycin propagates Akkermansia muciniphilia and reduces     Diabetes incidence in the  NOD mouse. Diabetologia 2012;55:2285-94.
  163. Hu Y,Peng J,Tai N,HuC,Zhang X.Wong FS, etal.Maternal  Antibiotic treatmentprotects offspring from   Diabetes development in non obese diabetic   mice by generation of tolerogenic APCs. J Immunol  2015;195:4176-85.
  164. Pinto E,Anselmo M,Calha M,Bottrill A,Duarte I,Andrew P, etal.The intestinal   proteome  of diabetic and control  children  is enriched with different  microbial and host proteins. Microbiology  2017;163:161-74.
  165. Gavin PG,Mullaney JA,LooD,Cao KL,Gottlieb PA ,Hill MM, etal. Intestinal   metaproteomics reveals the host – Microbiota interactions  in subjectsat risk for   Type1 Diabetes. Diabetes Care 2018;141:2178-86.
  166. Coppieters KT,Wiberg A,Von Herrath MG.Viral infections and molecular mimicry in Type1 Diabetes.APMIS 2012;120:941-9.
  167. Kramna L,KolarovaK,Oikarine S, PursiheimoJP ,Ilonen J,Smell O, etal. Gut Virome sequencing  in children with early  islet autoimmunity. Diabetes Care 2015;38:930-3.
  168. Zhao G, Vatanen T,Droit L,Park A, Kostic AD,Poon TW, etal. Intestinal Virome changes precede autoimmunity in Type1 Diabetes susceptible  chidren. Proc NatlAcad Sci USA 2017;114:E6166-75.
  169. Kim KW ,Horton JL,Pang CNJ,Jain K,Leung P,Isaacs SR, etal.Higher abundance of enteroViruses A species  in the gut of children with   islet autoimmunity.Sci Rep 2019;9:1749.
  170. Aw W,Fukula S.Towards the comprehensive understanding of the gut ecosystem via Metabolomic –based integrated omics approach .Semin Immunopathol  2015;37:5-16.
  171. Bridgman SL, AzadMR, Field CJ,Haqq AM,Becker AB,Mandhane PI, etal.Fecal short chain fatty acids variations by breastfeeding status in infants at 4 mths:differences in relative vs  absolute concentration.Front Nutr  2017;4:11.
  172. Trompette A,Gollwitzer ES,Yadava K,Sichelstiel AK,Sprenger N,Ngom –Bru C , etal. GutMicrobiota   metabolism of Dietary fibres influences  allergic airway disease  and haematopoiesis. Nat Med 2014;20:159-66.
  173. Benus RF,Van den Werf TS,Welling GW,Judd PA,Taylor MA,Harmsen HJ, etal. Association between  Faecalibacterium Prausnitzii  and  Dietary fibres  in colonic fermentation in healthy  human subjects. Br J   Nutr 2010;104:693-700.
  174. Singh N,Gaurav A,Sivaprak-asamaS,Brady E,Padia R,ShI H, etal.Activation of  Gpr 109a receptor  for niacin and the commensal  metabolites  butyrate suppresses colonic inflammation  and  carcinogenesis.Immunity 2014;40:128-39.
  175. Marino E,Richards JL,Mcleod KH,Stanley D,Yap YA,Knight J, etal. Nat Erratum:  GutMicrobial metabolites  limit the frequency of  autoimmune T cells and protect against Type1 Diabetes. Nat Immunol  2017;1841271.
  176. Songini M,Bernardinelli L,Clayton D,Montomoli C,Pascutto C,Ghislandi M, etal.The Sardinian IDDM Study:I.Epidemiology and geographical  distribution of IDDM in Sardinia during 1989-1994. Diabetologia 1998;41:221-7.
  177. CuledduN,Chessa M,Porcu MC,Fresu P,Tonolo G,Virgilio G, etal.NMR –based Metabolomic study of Type1 Diabetes. Metabolomics 2012;8:1162-9.
  178. Oresic M,Simell S,Sysi-Aho M,Nangto –Salonen K,Sappanen-Lasko T,Parikka V, etal.Dysregulation of lipid and aminoacid metabolism  precedes islet autoimmunity in children who later progress to   Type1 Diabetes.J Exp Med 2008;205:2975-84.
  179. Dolensek J,Rupnik MS,Stozer A. Structural similarities and differences between the human and mouse pancreas .Islets 2015;7:e1024405.
  180. Wandell PE,Carlsson AC.Time trends and gender differences in incidence  and prevalence   of  Type1 Diabetes in Sweden.Curr   Diabetes Rev 2013;9:342-9.
  181. ,Roep  BO,Atkinson MA,VonHerrath M.Satisfaction (not guaranteed):reevaluating  the use of animal models of  Type1 Diabetes.Nat Rev Immunol  2004;4:989-97.
  182. Manrique P,Bolduc B,Walk ST,Van der Oost J,De Vos  WM,YAoung MJ. Healthy human gut phageome. Proc NatlAcad Sci USA 2016;113:10400-5
  183. Sokol H,Leducq V,Aschard H,Pham HP,Jegou S,Landman C, etal. Fungal Microbiota dysbiosis in IBD.Gut 2017;66:1039-48.
  184. Koskinen K,Pausan MR,Perras AK,Beck M,Bang C,Mora M, etal.First insights into the diverse human archaeome :specific detection of archaea in the gastrointestinal tract,lung and nose and on skin.MBio 2017;8:e00834.
  185. Abu Ali GS,Mehta RS,Lloyd-Price J,Mallick H,Branck T,Ivey KL, etal. Metatranscriptomeof human fecal  microbial communities  in a cohort of adult men.Nat Microbiol 2018;3:356-66.
  186. Wilson MT,Hamios DL.The nasal and anus Microbiome in health and disease.Curr Allergy Asthma Rep 2014;14:485.
  187. Byrd BL,Belkaid Y,Segre JA.The human skin Microbiome. Nat Rev Microbiol 2018;16:143-55.
  188. Ma B,Forney LJRavel J. Vaginal Microbiome  :rethinking health and disease. Annu Rev Microbiol 2012;66:371-89.
  189. Niehaus L,Boland I,Liu M,Chen K,Fu D,Henckel C, etal. Microbial coexistence  through  chemical mediated interactions. Nat Commun2019;10:2052.
  190. Huang Q,Kahn CR,Atlindis E.Viral  hormones:expanding dimensions in endocrinology . Endocrinology2019;160:2165-79.
  191. Kahalehili HM, Newman NK, Pennington JM, Kolluri SK, Kerkvliet NI, ShulzhenkoN, etal. Dietary Indole-3-Carbinol Activates AhR in the Gut, Alters Th17-Microbe Interactions, and Exacerbates Insulitis in NOD Mice. Front Immunol. 2020; 11: 606441. Published online 2021 Jan 21. doi: 10.3389/fimmu.2020.606441
  192. Calabrese CM,Valentini A, Calabrese G. Gut Microbiota and Type1 Diabetes. Front Nutr. 2020; 7:617223.
  193. Verduci E,Mameli C,Amatruda M,Petiti A,Vizzuso S,Azzadi FE etal.Early nutrition and risk of Type1 Diabetes:the role of Type1 Diabetes. Front Nutr. 2020; 7:612377.
  194. Giampioli O,Conta G,Calvavi R,Micheli A.Can the FUT2 nonsecretory phenotype associated  with    Gut Microbiota increase the  children susceptibility for  Type1 Diabetes?A Minireview Front Nutr. 2020; 7:606171..
  195. Elhag DA,Kumar M,Khodore SA.Exploring the triple interaction between the Host Genome,the Epigenome and the Gut Microbiome in Type1 Diabetes.Int J Mol Sci 2021;22:125.