The Role of Microbial Metabolites in the Form of Modulators of Infant Gut Microbiome Along With Host - Microbial Crosstalk in Early Life: A Narrative Review

Kaur KK

Published on: 2023-11-04

Abstract

The   generation of infant gut microbiome is a key event impact the ecology along with function of the microbiome in addition to health of the host. Whereas, the   generation of the infant gut microbiome has caught attention for several decades, the  concentration on microbial metabolism in the gut as well as resultant  small molecules/ metabolites has been restricted .Nevertheless, technological in addition to computational  advancements are aiding researchers in evaluation the profile of myriad of metabolites in the infant gut, which would contribute in gaining insight with regard to the manner by  which these microbial metabolites guide structuring of microbiome community in addition to host microbial crosstalk. Earlier we reviewed generation of the Gastrointestinal Tract (GIT) in neonates particularly the hurdles met in feeding of preterm infants with associated complications like Gut, Brain &G-B Axis generation besides part of SIRT signalling in breast feeding &numerous articles on probiotics. Here we provide a narrative    review over the present information on the generation of the infant gut microbiota along with metabolism in the first year of life in addition to detail the manner by which these microbial metabolites are crucial for escalating insight with regard to crosstalk at the time of early life generational window.

Keywords

Metabolites; Infant; Microbiota; Health; Host; Metabolomics disease

Introduction

A germane simple   microbial community at the time of birth, generates into a variable in addition to complicated microbial ecosystem at the time of the   window of the early life [1], that possesses a key part in the postnatal   generation of the endocrine, nervous, along with immune systems [2]. Thereby the microbiome possesses considerable impact on the early in addition to long term health as well as generation [3]. Apart from impacting generation in addition to maturation of the immune systems of the host, the gut microbiota (GM) possesses numerous significant functions in the host inclusive of   degradation of consumed complicated carbohydrates, hampering pathogenic bacteria along with metabolic functions inclusive of     generation of Viamins, short chain fatty acids (SCFAS), as well as Bile Acids (BA) bioconversion [4]. Clarificationis present with regard to the significance of neonatal bacterial colonization for growth of a healthy subject in addition to possess the strength which microbiome has over its host at the time of early generational stages. To start with GM   constitution has been acknowledged to be in a stage of flux followed by later stabilization that results in their constitution being akin to that of the adult at 2.5-4yrs, along with akin properties of the gut in the adulthood [5]. At this early stage GM possesses greater susceptibility to modulation in contrast to adulthood [6], pointing to how significant the initial 1000 days of life   are (referring to the duration from conception till the 2nd birthday) portray a key opportune window with regard to ideal bacterial colonization. Interference  with microbial generation  at this  time  is  thought to be possessing long term inimical sequelae , which influence  normal immune as well as neurocognitive generation in addition to has  been correlated with escalated formation of diseases for instance,  asthma as well as metabolic syndrome (MetS)in latter part of life [7].

The GM might further be manipulated at this key juncture in an advantageous way. Approaches with regard to attainment of early   ideal microbial at the time of this generational opportunistic crucial   window might be contributing to the avoidance as well as the treatment of diseases. Attempting manipulation of the early microbiome are inclusive of utilization of  Prebiotics, Probiotics or combination therapy of both in  the form of synbiotics [8] or  posbiotics [9].The maximum attractive outcomes of   Probiotics havebeen observed in  the avoidance as well as the treatment of diarrhea along  with necrotizing  enterocolitis (NEC) in case of preterm infants[10]. Pharmabiotics inclusive of microbially   generated bacteriocins, bacteriophages as well as SCFAs have invoked substantially   enhanced interest with regard to probable applications regarding microbiota manipulation [11]. Moreover, in recent years  a switch has  further been made regarding expansion of research further than taxonomic constitution with the utilization of  16S rRNA    gene  sequencing  as well as concentrating  on gut microbial  working as well as activity with the utilization of  whole metagenomic sequencing(WMS) along  with metabolomics. Despite,    more expensive, WMS in contrast to 16S rRNAgene sequencing   gives understanding regarding encoded by the    genes existent in the microbiome. WMS further promotes examination of the community structure at considerably greater as well as dependable resolution of taxonomy further than levels of genus for examination of the species (spp) in addition to subspp 12]. Additionally, metabolomics is a strategy which is complementary regarding determination of themetabolites in the gut obtained from the diet,host or microbiota(together referred to as  Gut microbiome)[13]. Advancements in informatics for instance machine learning, have further yielded provision of gaining greater understanding along with promoted interpreting functional microbiome research with greater meaning [14].

Earlier we reviewed generation of the Gastrointestinal Tract (GIT) in neonates particularly the hurdles met in feeding of preterm infants with associated complications like Gut, Brain &G-B Axis generation besides part of SIRT signalling inbreast feeding &numerous articles on probiotics [15-17].

Here we have described in brief the factors   which impact GM in the early life, detailed present publications with regard to diet generation of the infant gut microbiome in addition todescribe the manner   by which microbiota obtained metabolites might be crucial regarding early life microbiome as well as hostgeneration.

Methods

We conducted a narrative    review utilizing the Pubmed search engine on, using the MeSH terms Bile Acids(BA );Gut microbiota( GM); Delivery modes; vaginal  delivery; caesarean section;  Early Life; Breast milk ;human milk oligosaccharides (HMOs); Bifidobacterium; Formula milk; Enterobacteriaceae/ Clostridium; Microbial metabolites ; 2’fucosyllactose(2’FL) ; prebiotics supplementation; long chain fructoligosacchharides (lcFOS), HMOs supplementation  Farsenoid X receptor[FXR]; Takeda G protein coupled (GPC) bile acid receptor 5(TGR5);GPCR;GPR41/GPR43;HCA from1990’s   till datein October  2023 .

Results

We found a total of 7050 articles out of which we selected 144 articles for this review.No Meta-analysis was done.

Factors Impacting Gut Microbiota (GM) In Early Life Microbial Metabolites 

Non Nutrition Factors: Delivery modes possess a substantially significant impact on the GM constitution, with the exposure of the infants to variable kinds of microbes correlated with maternal gut along with vaginal tract [18]. Infant passing via the birth canal gets exposure to the maternal vaginal as well as faecal microbiota, which aids in microbial along with metabolic variations in infants gut [19]. Lower segment caesarean section (LSCS) delivery   results in dysfunctional shift of some maternal strains that leads to escalated enrichment of probable pathogenic microbes with the absence   of advantageous taxa for instance Bifidobacterium[20]. Other factors impacting  the generation of the infants  gut microbiome revealed  are inclusive of     i)Gestational age,ii)host  genetics,iii)feeding patterns,iv) geographical area in addition to v) perinatal exposure to antibiotics[21]. Gestational age portrays a significant factor in the generation of the GM with the preterm birth being correlated with a diminished diversity along with postponed colonization of Bifidobacterium in contrast to full term infants [22]. Exposure to antibiotics in early life has been displayed to lead to induce escalated enrichment of members of Enterobacteriaceae family as well as reduction in enrichment of Bifidobacterium[23]. Exposure to antibiotics has further been correlated with the generation of the metabolic in addition to inflammatory diseases in the latter part of life [24]. Recently it has been corroborated that there is presence of breast milk microbiome which works in the form of a source of the advantageous microbes towards the infants [21, 25]. Furthermore, acquisition of microbes which result  in colonization of the infants gut    takes place  from the environment  are inclusive of   siblings,pets as well as the hospital[3,26].

Breast Milk: Human breast milk possesess the capacity of taking   care of all nutritional needs for the growth, health along with generation of the infant [27]. Breast milk is constituted   of necessary nutrients for instance casein, fatty acids (FA), in addition to lactose apart from micronutrients inclusive of immuoglobulinA, immuoglobulinG as well as immuoglobulinM, calcium along with Vitamin A [28]. Moreover, breast milk is a rich source of a wide variety of bioactive constituent’s for instance human milk oligosaccharides (HMOs). HMOs possess over 150 acknowledged complex glycans of which the maximum frequent is 2’fucosyllactose (2’FL) which is implicated in 20-30% of all HMOs [28,29]. Digestion of the HMOs does not take place in  the upper gastrointestinal tract(GIT), as well as  their breakdown takes place in  the colon  by particular bacteria  meant for HMO breakdown ,mainly Bifidobacterium  for breaking down HMO with efficacy, into lactate as well as acetate having  probable advantageous host health actions[29]. The capacity of certain species (spp’) of Bifidobacterium to breakdown with effectiveness labels them in the form of dominant members of the microbiota in case of breast fed infants. The liberating status of host is dependent on the active or inactive copy of the fucosyl transferase-2(FUT2) gene, estimates the maternal capacity of generating HMOs [30]. The non liberators have been correlated with reduction in Bifidobacterium as well as enrichment of Bacteroides in addition to lesser microbial variation in their infants GM [31, 32]. The lack/ reduction of 2’FL in non-liberator women have probable short along with long term repercussions regarding health along with generation [33].

 Formula Milk: Constitutional assessment of the infants has illustrated changed community structure in case of formula fed   in contrast to breast fed infants [34]. Greater enrichment of probable pathogenic bacteria for instance Enterobacteriaceae as well as Clostridium in addition to antimicrobial resistance genes has been found in case of formula fed infants in contrast to purely breast fed infants [5, 34,35]. On the other hand feeding breast milk is correlated with lesser microbial variations, with dominance of HMO using Bifidobacterium  spp in addition to saccharolytic  fermentation in contrast to formula feeding [36]. In view of greater quantities of proteins in formula milk, infants fed formula milk might illustrate greater   proteolytic metabolism in the gut [37]. Proteolytic microbiota obtained metabolites have been correlated with inimical cardiovascular as well as metabolic health sequelae in adulthood [38-41], as described in greater details later. Nevertheless, if alterations in colonic fermentation found in case of formula fed infants impact the risk of generation of the diseases in latter part of life needs clarification. Nevertheless, numerous recent infant formula have received supplementation with prebiotics for simulation of the advantageous actions of HMO in the breast milk. Prebiotics supplementation has usually been done with inclusive of long chain fructoligosacchharides (lcFOS), short chain   galactooligosacchharides (scGOS), in addition to HMOs for instance 2’fucosyllactose (2’FL) as well as Lacto-N-neitetraiose(LNnT) [42].

Weaning & Further: At the time of the switch  towards  solid  foods canonically amongst  4-6 mths , alterations take place  in  the enrichment along  with the variations in  the infants GM. Bacterial  spp which have adapted  to milk for instance Bifidobacterium longum subspp infantis canonically undergo reduction at this  time period[43]. In view of the escalated ingestion of the protein as well as dietary fiber at this time period microbes possessing the capacity of digesting the greater complicated nutrients get enriched for instance Lachnospiraceae, Bacteroidacea, in addition to Ruminococcacea [44,45]. Maturation of microbiota take place persistently further than 5 years as well,where  the children have attained the variation, stabilization, in addition to adult like organizations, however with the lesser enriched community in contrast to the adult microbiota [46].

Malnutrition: Food portrays  a main factor  implicated  in  the provision of   shape  to the GM in early life period.This further becomes apparent in case of those  not receiving enough nutrition at the time of  infancy as well as early childhood .An immature gut microbiome   generation takes place in case of infants as well as children who are malnourished[47], that might result  in dysfunctional child  growth[48]. This disruption of microbial succession in the children who are malnourished might aid in lifelong   aberrant growth in addition to generation [reviewd separately ref 49]. Interestingly, a recent3mth long study with involvementof intervention illustrated microbiota guided complementary food paradigm in contrast to the present ready for use supplementary food   facilitated weight accrual in children presenting with moderate acute malnutrition amongst 12-18mths of age [50]. This study pointed that improvement of weight accrual might be modulated by a greater full healing of the GM [74]. Corroborating   this posit,a placebo  controlled trial in malnourished children who were 2-6 mths of age  from Bangladesh, having robust malnutrition observed that supplementation with Bifidobacterium longum subspp infantis facilitated weight accrual which was correlated with  reduction  in quantities of intestinal inflammatory markers[51]. Worldwide in 2020,149 million children less than5 yrs age  were determined to be  stunted [52], highlighting the requirement  of the gaining insight with regard to immature gut microbiome in case of children who are malnourished.

Colonic Fermentation Along With Microbial Metabolites in Early Life: The transitory period from  an  only source of the milk to a wide variety of  foods[53] apart from  shaping gut microbiome constitution[1,5,54], further results  in shaping of   gut microbial  metabolism  in early life that leads to formation  of numerous small  molecules(alias metabolites)(Figure1)(rev ref 55] .

Figure 1: Courtesy ref no-55-Gut microbiota-derived metabolites in early life. Gut microbial metabolism changes in early life with the progression in early nutrition from breastfeeding to solid foods concurrent with a progression in microbiome and metabolome diversity. During breastfeeding, the dominance of the infant gut by human milk oligosaccharide (HMO) degrading Bifidobacterium species results in high levels of lactate and acetate, as well as in aromatic lactic acids (i.e. phenyllactic acid, 4-hydroxyphenyllactic acid and indolelactic acid). With the progression in diet, the dietary complexity increases and more indigestible proteins and fiber end up in the colon of the child. Consequently, colonic fermentation changes resulting in the formation of short-chain fatty acids (SCFA, i.e. acetate, propionate and butyrate) and gases (i.e. hydrogen and methane). Furthermore, proteins are degraded into amino acids, which are fermented by the resident gut microbes into branched SCFAs (i.e. isobutyrate, isovalerate and 2-methylbutyrate), amines (i.e. histamine, dopamine, tyramine, γ-aminobutyric acid (GABA), tryptamine), as well as aromatic acetic and propionic acids (e.g. indoleacetic acid and indolepropionic acid).

Colonic Fermentation of Carbohydrates:  The maximum assessment of microbiota obtained metabolites has been of the short chain fatty acids (SCFAs). Generation of  SCFAs  takes place as a result  of colonic fermentation   of  HMOs, dietary fibers in addition to proteins, further acknowledged  to be impacting  human metabolism in multiple manners through activation of variable  host  receptors in separate organs[56]. SCFAs further work  in  the form of energy substrates  for the resident  gut microbes  in addition to human cells with butyrate  acting in  the form of a primary energy substrate  for  colonocytes[57], along  with propionate working   in  the form of substrate  for gluconeogenesis in  the intestine[58], as well as liver[59]. With regard to adults, the main SCFAs are acetate, propionate, along with butyrate are canonically observed in a 3:1:1 ratio,while succinate in addition to lactate apparently work  in  the form of intermediates, thereby possess variable quantities . Nevertheless, in early life alterations in SCFAs profile takes place paralleled with child’s   propagation in dietary as well as microbiome variability [60,61]. At the time of breast feeding(early phase), the properties of  SCFAs are lesser acetate as well as greater  succinate at the time of complementary feeding(middle  phase), with greater lactate,  pyruvate as well as  formate followed by subsequent to stoppage of breast feeding(late   phase)   by greater propionate, along  with butyrate[60].The greater enrichment of lactate, pyruvate as well as  formate estimated in  the infants gut at the time of early phase portrays  a lesser   generated microbiome in view of these metabolites with regard to  a well generated gut microbiome, canonically would have  been transformed into other metabolites[56,62]. In contrast to infants that  are not breast fed,totally breast fed infants   have  been displayed  to possess lesser absolute quantities of  full SCFAs inclusive of  acetate, propionate, butyrate,valerate,isobutyrate,   isovalerate; however greater  quantities of  lactate at4mths age[63]. Bifidobacterium are substantially enriched in breast fed infants[64], in view of their capacityof  metabolizing HMOs[65], that leads togeneration of acetate in addition to lactate[66]. Nevertheless,   stoppage of breast feeding as well as formation of the gut   microbiome, escalated butyrate generation takes place in the infant’s gut [60]. Although, the greater dynamics in SCFAs   are found in early life which have probable significance in the form of signalling molecules, the association amongst SCFAs, gut   microbiome along with carbohydrates substrates   are restricted. Moreover, with greater than 200 HMOs kinds isolated in human breast milk in addition to greater differences in HMOs profiles amongst mothers as well as   right through lactation [67]. Furthermore,it needs to be worked out,the  these variations along  with the manner, in which  HMOs profiles influence the infants gut microbiome metabolism.

Colonic Fermentation of Proteins: Studies with regard to metabolites of the GM in case of early life are basically restricted regarding SCFAS [66,68]. Nevertheless, with the acknowledgement of  escalated ingestion of the dietary fiber in addition to proteins at this  time period with the complementary diet[53,69], that is correlated with escalated gut microbes variations[68] in addition to capability  of transportation of  amino acids at the time of infancy[5,54], numerous  microbial obtained  metabolites from dietary fiber as well as proteins[3,5 ,6,70,71]are presumably undergoing alteration with this dietary transit. For instance,it got recently, revealed that HMOs breaking down Bifidobacterium  spp transform aromatic amino acids(for instance phenylalanine, tyrosine along  with tryptophan) into aromatic  lactic acids( indole lactic acid, phenyl lactic acid, as well as 4 hydroxy phenyl lactic acid ) in  the infants gut[88]. Paralleled with the breast fed infants having dominance  of Bifidobacterium[68], aromatic  lactic acids have  been observed to be in greater  quantities in  faeces of  breast fed infants in contrast to  formula fed  infants[61,72] as well as infants which are  weaned[64], that highlights the robust crosstalk amongst early life nutrition in addition to impacting the quantities of   microbial  aromatic amino acids catabolites in early life. Other instance of microbiota obtained metabolites in early life are the branched chain fatty acids (BCFA)[ isovalerate,2- methyl butyrate). These  BCFA  portray  enhanced  proteolytic fermentation in view of their origin  from  branched chain amino acids (BCAA)[ valine,leucine along  with  isoleucine][13], which are held responsible for insulin resistance(IR)[73].The quantities of escalated proteins along  with the  amino acids possess the  part in the dominance  of the quantities of BCFA in view of observations  in a study with regard to breast fed infants  had  significantly   lesser quantities of faecal BCFA in contrast to  those fed  2 infant formulas with the remarkable hydrolyzed  proteins along  with the  amino acids respectively  at the time of 1 as wellas 2mths age[74]. Canonically infant’s formula possesses greater quantities     of proteins along with the lesser amino acids in contrast to breast milk [75], pointing that the greater fermentation of proteins takes place in the colon of the formula fed infants. In agreement with this,a study in case of   newborn rhesus monkeys observed that  by reduction of  quantities  of proteins of the formula, quantities  of urinary microbial obtained proteolytic substances (for instance  3- indoxylsulphate, 4 hydroxy phenyl acetic acid as well as 2 hydroxy isobutyrate) could get diminished[76]. Furthermore, quantities of BCFA have further been correlated with differences in breast milk lipids as well as HMOs constitution [77]. Moreover, BCFA have been  displayed to escalate in  the gut of the    infant with time that is   independent  of early feeding (breast feeding vs formula feeding)[37,60,], pointing that the absence of HMOs subsequent to propagation towards solid foods facilitates  a switch towards amino acids fermentation in  the gut of the infant. Further biogenic amines ( for instance    cadaverine,tyramine) have been  displayed in  greater quantities  in faeces   of the formula fed  infants in contrast to breast fed infants[78]. These   variations might takes place in view of  variations in gut microbiome constitution, in view of infants formula correlated genera   inclusive of  Streptococcus in addition to Enterococcus[79],both of these are generators of putrescine, cadaverine, along  with tyramine[80].Their isprevalence of one more commensal in case of  early life  once propagation towards solid foods takes place in children namely  Ruminococcus gnavus[81] that further possess the  capacity  of forming  tryptamine  in vitro[82]. Nevertheless, there is need for assessment if R. gnavus truly results in formation of tryptamine in the infants gut. Actually numerous microbial proteolytic obtained metabolites correlated with inimical sequelae on  the cardiovascular in addition to metabolic health in adults, for instance  trimethylamine-N-oxide (TMAO , obtained from carnitine,betaine    along  with choline)[38], imidazole propionate( obtained from histidine)[39], phenyl acetyl glutamine(obtained from phenylalanine )[40], p-cresyl sulphate as well as  indoxyl sulphate( obtained from tyrosine as well as  tryptophan)[41], continue  to be underevaluated regarding early life . These metabolites   would be anticipated to get initiated on transitionof children to adult’s kind solid foods. Nevertheless, least number of studies have investigated the influence of complementary diet on the infants gut microbiome [48].

Colonic Fermentation   of Lipids Along With Bile Acids: Dietary  fat further gets metabolized by microbes present in  the  GIT.Via breast milk in addition to in  certain cases of formula milk, provision of essential  fatty acids linoleic  acid(LA) in addition to α-linolenic  acid (ALA) along  with the long  chain polyunsaturated chain fatty acids (LC-PUFA’s): eicosapentaenoic acid (EPA), arachidonic acid(AA) docosahexaenoic acid (DHA, omega  3 fatty acids )[53]. Apart from  possessing direct actions in  the  gut microbiome constitution in addition to actions[83], the PUFA’s possess the  capacity  of  getting metabolized by    colonic microbes  into hydroxy  fatty acids(FA),oxo fatty acids, along  with the conjugated fatty acids in addition to partly saturated transfatty acids, in  the form of intermediates[84]. Intriguingly, particular Bifidobacterium strains have  been observed to possess the  capacity  of transforming LA  to conjugated linoleic  acid(CLA)[ 84,85]., that has  been correlated with a range of systemic health facilitating actions [86]. Despite, the generation along  with significance of  CLA in  the infant’s  gut  are uncertain  at  present,one more LA obtained substance 12,13-  di hydroxy  9-Z octadenoic acid (12,13-  diHOME) have  been pointed to hamper immune tolerance in early life [87].This metabolite was recently revealed  to be generated by early life members for instance  B. bifidum in addition to E. faecalis strains[87]. Moreover ,a   recent subsequent randomized  controlled  clinical trial(RCT), revealed that infants  receiving formula milk possessing HMOs (2’fucosyllactose along  with LNnT) possessed diminished faecal quantities of  2 di hydroxy fatty acids(12,13-  diHOME as well as 9,10 di hydroxy  12-Z   octadenoic acid [9,10- diHOME] ) in contrast to  infants receiving control  formula milk without HMOs, that pointed to HMOs stimulated changes in  the microbiome might influence microbial transformation of fatty acids[88]. Thereby greater studies  are needed for evaluating intestinal FA metabolism  in the infants  gut, that  would probably  be  on  the consumption of  FA quantities, the effectiveness  of absorption in  the  small intestine in addition to the  distinct gut microbiome constitution.

On consumption of fat, liberation of bile acids takes place into the small intestine for contributing in digestion of lipids. Bile acids comprise the primary constituent   of bile  apart from  gets conjugated with glycine(main in humans- 75%) along with taurine respectively [89].Whereas reabsorption  of 95% of bile acids takes place in  the  ileum along  with  gets   transported back to the liver through the portal vein,   while nonabsorbed stay back in  the colon where they get chemically converted to secondary  bile acids[90]. Deconjugation of bile acids by microbes leads to diminished reuptake in the small intestine that influences the pool of bile acids which enter the colon [90]. Additionally, microbial modifications of bile acids influences  their    chemical structure which results  in changed signalling through host receptors  inclusive of  G protein coupled (GPC) bile acid receptor 5(TGR5), Farsenoid X receptor[89], that might have main influence on host   metabolism described by us   in detail [ 91]. Concurrently with the   succession of the gut microbial  community in early life , generation of   intestinal  bile acids takes place as per both the microbiota stage  along  with that of  the nutritional stage .In a longitudinal study performed over  10 healthy Japanese individuals   it was observed that at the time of the first 6mths of early life infant’s with the dominance of Bifidobacterium spp’ possessed  the  greater faecal  quantities of unconjugated  primary bile acids in contrast to infant’s with the dominance of  Enterobacteriaceae species  possessed  the  greater faecal  quantities of conjugated  primary bile acids[92]. Actually bile salts hydrolase activity that is  implicated in  the catabolism of deconjugation of conjugated  bile acids at the time of  breast feeding  is  more frequent in Bifidobacterium[93], pointing that Bifidobacterium is a  crucial  actor for deconjugation of conjugated  bile acids at the time of  breast feeding. Nevertheless,on  propagation towards   solid diet, escalation  of dietary energy  quantities takes place [53] enhancement of dietary complicated nature takes place[68]. Simultaneously,full excretion of faecal bile acids in addition to faecal  quantities of secondary bile acids   enhance[61,92]. Additionally, diminished faecal  quantities of primary bile acids in addition to   enhanced faecal  quantities of secondary bile acids have  been continuously observed  subsequent  to weaning[92,94]. Variations in faecal bile acids have further been found on contrasting breast fed infants as well as formula fed infants with breast fed infants   revealing enhanced quantities of secondary bile acids [41].  Lastly, in mice,a study illustrated that  oral delivery   of bile acids to  newborn mice augmented  postnatal  gut microbiome maturation[95], pointing that propagation  of bile acids profiles   at the time of infancy guides maturation of the    gut microbiome.

 Microbial Metabolites   in The Form of the Structuring of the Microbiome Communities: Gut microbial transformation of dietary substrates into metabolites portrays a probable controlling modes by which particular Gut microbes influence the broader gut ecosystem (Figure 2).

Figure 2: Courtesy ref no-55-Microbial metabolites as modulators of the infant gut microbial ecosystem. An illustration of the ways in which microbial metabolites are thought to impact the development of the gut microbiome ecosystem during infancy. Human milk oligosaccharides (HMOs) of breast milk are likely to enter the distal gut and be degraded by HMO degraders (for example Bifidobacterium Bacteroides, Ruminococcus and Akkermansia). Among the HMO degraders, particular Bifidobacterium species are important as they are specialized in HMO degradation. Some Bifidobacterium species (mainly B. longum subsp. longum, B. bifidum and B. breve) degrade HMOs extracellular, whereas other Bifidobacterium species (mainly B. longum subsp. infantis) take up the HMOs via ABC transporters and metabolize them inside the cell. The sharing or lack of sharing of nutrients (both carbon sources and micronutrients) have consequences for cross-feeding and competition in the infant gut microbiome affecting the microbial community structure. As the child transitions from a milk-based diet to a solid food-based diet, the microbiome diversity increases and the cross-feeding networks change. Other primary degraders of dietary fibers (typically belonging to Lachnospiracea, Bacteroideacea, Clostridiacea) take over and replace the Bifidobacterium species. Generation of specific microbial metabolites may be key for colonization resistance toward pathobionts when microbiome diversity is low. The pathobionts may be excluded or kept low in abundance through low intestinal pH (due to short-chain fatty acids, SCFAs), through direct antimicrobial activity such as bacteriocins, which are antimicrobial, or simply through nutrient limitations.

Specifically HMO obtained metabolites generated by those which are implicated in primary HMO breakdown guide crossfeeding networks which promote of other early life taxa in addition to interspecies crosstalk [96,97]. Infants possess very poor capacity  of digesting HMO; nevertheless  are the preferred growth substrates for some Bifidobacterium spp’  as well as strainswhich possess the  capacity  of enzymatically breaking down complicated dietary constituents  inclusive of   LN(n)T ,fucosylated as well as  sialylated  structures[98].  in addition to  Bifidobacterium, Bacteroides as well as Ruminococcus,with recent  inclusion of Akkermansia  muciniphilia   have further  been illustrated to possess the  capacity   of  breaking down complicated carbohydrates despite in maximum instances this is   correlated with enzymatic cleavage of sugar  side chain in addition to associates with correlated mucin breakdown pathways[99].Some  Bifidobacterium spp’ apparently possess restricted influence  on  the   broader  gut ecosystem  structuring in view of their selfish digestion of  HMOs. This specifically holds good for Bifidobacterium longum subspp infantis  strains where they are actively transported (through Apical Sodium Dependent Bile Acids Transporters ([ASBT])  as well as  internalize  full(with integrity) HMOs  for ensuring intracellular degradation ,with the idea of utilization of the  monosaccharide metabolites  for their  own   personal cellular events, instead of sharing amongst  other early life microbes [100]. Other species   for instance B.bifidum,Bifidobacterium pseudocatenalutum, Bifidobacterium longum subspp infantis as well as B.breve further perform  extracellular hydrolysis  of complicated HMOs followed by import along  withmetabolizing the resultant mono, as well as disaccharides. Thereby these strains possess a significant milestone function at the time of early life by provision of simpler metabolites with regard to crossfeeding [101]. Metabolites possessing  the  capacity   of  getting utilized  by other secondary microbes  for breaking down are  inclusive of SCFAs lactate & acetate, succinate, pyruvate ,formate, fucose as well as  1,2, propanediol(1,2PD)[60,102]. Earlier,work  has  pointed that amongst Bifidobacterium mini ecosystem ( from separate infants) usually there  is presence  of             milestone member  which actively leads to degradation various HMOs, that results  in  provision of crucial metabolites for instance fucose, galactose, acetate as well as N acetyl glucosamine  that    gets further utilized  by other  non HMOs degrading strains[103]. Other researchers  corroborated this posit illustrating  that at the time of growth on HMOs crossfeeding crosstalk amongst numerous Bifidobacterium strains escalate total  cell numbers, that was correlated with generation of formate, acetate, 1,2, propanediol as well as lactate at the time of cocultivation[97].A  study has recently  pointed  the manner  by which timing  of sugars in  the  infant  gut shape the broader Bifidobacterium ecosystem,  having  dominance of B.bifidum, as well as Bifidobacterium longum subspp infantis via  hampering priority actions  dependent on their capacity  of digestion of HMOs, whereas B.breve  takes advantages from the promoting actions in addition to dominting by use of fucose(result  in alignment with the profiles found in metagenomic outcomes)[104]. These studies emphase the  significance of crossfeeding  alias  ‘syntrophy’  for Bifidobacterium  whose definitions  is mandatory symbiont metabolism where without total mini ecosystem,all or combined strains do not possess the  capacity  of survival without each other .This totalistic strategy with regard to  use of HMOs might be pointing   towards extra events by which numerous Bifidobacterium spp’ as well as strains  exert dominance in  the breast fed infants gut in addition to are successful  in outcompeting other bacterial taxa. Actually, generation of acetate as well as lactate  by Bifidobacterium (in reaction to  the HMOs metabolism) is correlated with reduction of  intestinal pH , that is further associated  with  alteration in microbial  constitution, for instance reduction of Enterobacteriaceae,thus escalating colonization resistance in  the early life gut[105].  Paralleled with that in vitro experiments have observed that SCFAs possess the capacity of directly hampering the growth of pathogens from the Enterobacteriaceae family inclusive of Escherichia Coli (E.Coli)  Salmonella   enteridis ,Klebsiella pneumoniae,in a pH based  way[106] .

Watching further than Bifidobacterium HMOs obtained metabolites further crossfeeding   of other early life taxa. In vitro studies have further illustrated that growth ofLimosilactobacillus reuteri gets escalated via 1,2PD generated by B.breve UCC22003 [102]. Furthermore, numerous studies have evaluated the influence of crossfeeding on variety of butyrate    generating bacteria acknowledging their crucial part in facilitating health. In particular , butyrate    generating bacteria possess the  capacity  of use of acetate as well as lactate in addition to HMOs obtained monosaccharides , in  the form of carbon sources , that  work in escalating their quantities in  the infants gut. For instance, Anaerostipes caccae has  earlier illustrated  to grow  in coculture with just Bifidobacterium longum subspp infantis, resulting   in butyrate generation[107].Extra SCFAs generated  by eubacterium hallii (in reaction to  the (fucoslyated) HMOs degradation by B.breve as well as/or B. longum subspp infantis aided use of  L-fucose( for butyrate generation)  in addition to 1,2PD uptake ( for  propionate generation)[96], despite propionate has yet to be illustrated in  the infants gut. The accessibility  of L-fucose further corroborates growth  of other microbes  which encode Fuc  catabolic pathways for instance  B.thetaiotaomicron as well as Akkermansia  muciniphilia however these microbes further possess the  capacity  of  enzymatic cleavage of fucose from fucoslyated HMOs as per their fucosidase  action[108].

Noticeably, certain pathogenic bacteria have evolved approaches of utilization of fucose with regard totheir benefit inclusive of Salmonella as well as C. jejuni , that in  the   latter  instance apparently has enrichmentin case  of breast fed infants in contrast to those on formula[109]. Thereby the accessibility of different metabolites in the infants gut might be possessing inimical as well as advantageous actions based on ecological situations.

In certain subjects there might be  competitive crosstalk through generating metabolites acknowledged to  generate these antimicrobials hamper   metabolic reactions as well as growth of other microbial members for instance Hydrogen Sulfide (H2S), or SCFAs (reduction of pH)or through generation of metabolites possessing  direct  antimicrobial  actions for instance  bacteriocins Bacteriocins portray ribosomally generated anti-microbial peptides that possess actions against other bacteria in addition to possess a narrow spectrum (by target akin species) or wide spectrum (across variation ofgenera), with the resistanceof of host bacterium [110].  Numerous early life  microbiota members inclusive of Bifidobacterium ,are acknowledged  that might be correlated with their   capacity  of modulating    the  broader microbiota as well as hampering the overgrowth  of other  probable  pathogenic bacteria that  otherwise might be  inimical for the infant[111].The gene clusters needed for biogeneration are apparently strains particularly earlier work  has pointed that bacteriocins  bifidin(generated  by B.bifidum, NCDC1452) possesses actions againsts  wide variety of gram positive as well as  gram negative microbiota members inclusive of Escherichia Coli (E.Coli) along  with Staphylococcus aureus[111], thatare frequent early life  microbiota members. Other Bifidobacterium correlated bacteriocins are present; nevertheless, these have illustrated actions against particular food borne pathogens inclusive of Salmonella spp  along  with Bacillus cereus , thereby they might not be germane with regard to first  12 months of early life . Recently B. longum subspp infantis LH _664   been illustrated  to  generate a  narrow spectrum  bacteriocin(ct presumably ka  Bifidococcin _ 664) that   not influencing Lactobacillus as well as Enterococcus strains actually possesses     distinct actions  against a plausibly significant pathosymbiont in  the (preterm ) gut Clostridium perfingens[112].The capacity  of these active peptides in targeting particular bacteria might be of  considerable  significance with regard to early life disturbances( for instance preterm birth  caesarean section as well as /or antibiotics therapy)when quantities of  pathosymbionts might be  greater, acknowledging that lack of these vital metabolites/peptide modulating  factors which would otherwise  be  generated  by Bifidobacterium that  is eliminated in  these conditions . Nevertheless, it is not certain at this stage   if dietary (for instance HMOs) environment in the gut has a part in the shaping of the    generation of these peptides, with requirement of more work for getting insight on these associations.

 Once extra complicated dietary constituents  get  introduced (specifically plant obtained) at the time ofweaning remarkable expansion in crossfeeding networks in view of complicated environment of metabolites generated  by those which are implicated in  primary breakdown[101].This is in view of broader  range of glycoside hydrolaseencoded across variable taxa(Specifically in  the Bacteroides genus,maximum of which work in  the form of primary  degraders ) thataids in broader  range of bonds amongst  monosaccharide units meant for breakdown, thereby aiding  these simple sugars to gain entry  into  the variable pathways for yielding energy[ adenosine triphosphate(ATP)] regarding cellular reaction in addition to growth[113].Endproducts   of fermentationof get further broken down  by crossfeeders  that usually leads to generation  of SCFA, with escalating generation  of butyrate as well as propionate  in contrast to acetate ; dominant SCFAprofile found at the time of milk dependent   feeding alone[114].Extra crucial constituents  which can be  generated in view of protein-fermentation  are amino  acids, which  possess a  central  part with regard to  metabolism for numerous  bacteria(enzymes co factors)guiding  growth in addition to alterations in motility,biofilm  generation as well as expression of  virulence factors[115]. Additionally,  variations at the  strains level with regard to     Vitamin B biogeneration, that is the need  of the   host, in addition to other strains who do not possess the  capacity  of de   novo’’ generation  of  these crucial metabolites [116]. For instance variable early life taxa are anticipated as well as have been validated experimentally,to generate Vitamin B 1 inclusive of Lactobacillus spp, Clostridium spp, Bifidobacterium spp[117]. These strains are anticipated to possess a crucial partfor other spp in addition to strains who do not possess the  capacity  of Vitamin B 1 production( for instance Faecalibacterium spp) emphasizing the crossfeeding as well as competition with regard to  this imperative metabolite in  the infants gut[118]. Clarification,exists  that this  time  duration portrays a substantially dynamic time  duration of ecosystem production with the fluorishing crossfeeding networks  getting  generated as well as like other poorly generated ecosystems might be specifically    sensitive to diet obtained disturbances[119]. Nevertheless, Clarification of  corroborative proof  on  the long  term actions  at this  time  duration are needed through  longitudinal cohort or experimental studies . Oliphant as well as Allen-Vercoe have provided a detailed expansive review on this topic [114].

Lastly, in the Reference of spatial terms of these probable metabolites crosstalk need to be taken into account .It is clearer that the quantities along with diffusion capacity of crucial metabolites    be anticipated   to possess the maximum considerable influence on the ecosystem structure on the microbes intricately placed witheach other. Nevertheless, acknowledging the complicated geography of the   gut inclusive of total length,mucus layers in addition to microstructure  like crypts,it is probable that the  maximum studies conducted  thus far might  not fully reproduces these spatial  contexts. Thereby extra model system in the form of organoids as well as mice might be needed to precisely determine the germane modes of microbial metabolites in the form of modulators of the infants gut microbiome.

Microbial Metabolites in the Form of the Modulators of the Host Microbial Interactions in Early Life

Acknowledging  the dynamics  of microbial metabolites   in  the gut along  with their absorption into  the circulation[120], these microbiota obtained metabolites apart from shaping the  gut microbiome ecosystem, they further are correlated with modulation of the  host   microbial   interactions  in early life in view of they might be connecting with host  cells through host  receptors in  the intestine in addition to  far off organs (Figure3)[13,121].

Figure 3: Courtesy ref no-55-Dynamics of selected signaling metabolites in the infant gut and their interaction with host receptors. The figure illustrates the change in relative abundances of known key microbiota-derived metabolites generated from saccharolytic84 and proteolytic fermentation [84,88] as well as from bile acid metabolism132 in the infant gut. The metabolite profiles change as the infant goes from breastfeeding to solid foods concurrent with a maturation of the gut microbiome from a less to a more diverse microbiome. The main host receptor targets for each of the signaling metabolites is indicated to the right. As indicated by the arrows, some metabolites can bind several different host receptors.

Nevertheless, paucity of  studies have performed assessment of luminal as well as circulation quantities of the metabolites   in   early life.This has   significance in view of  considerable gap  to find out if quantities of the metabolites are  within the range  of the dose response curve  for the  specific metabolite on  the host receptor. Here brief description of 4 fields where microbial metabolites might possess crucial part in early life is given (Figure 4).

Figure 4: Courtesy ref no-55-Gut microbiota-derived metabolites as mediators of host-microbial cross-talk in early life. The microbiota-derived metabolites can impact host development in various ways as they activate different host receptors. For example, microbiota-derived metabolites can stimulate the intestinal immune system (affecting release of cytokines), enteroendocrine cells (affecting release of gut hormones), and enterochromaffin cells (affecting release of serotonin). Subsequently, the secreted molecules can affect the intestinal barrier and immune function, the intestinal nervous system with effects on intestinal motility and the gut-brain axis, and potentially the host metabolism and inflammation throughout the body upon absorption and circulation through the portal vein.

Microbial metabolites along with Intestinal   Barrier Function Reprogramming   Despite,a  robust barrier is  believed to  be necessary  for host health, at the time of early life,a  balance  is needed;aiding  crucial  microbial metabolites in crossing in addition to   start  immune programming  in  the underlying mucosal layer along   with far off  as well as  systemic  immune regions. Butyrate represents the   major fuel by intestinal epithelial cells (IEC) with regard to    growth [122]. Nevertheless, quantities of butyrate  are  germane low at the time of  milk feeding, however it gradually escalates  over the weaning    period as well as further[84], in view of the expansion of butyrate generating bacteria acknowledging  the complicated dietary  milieu [84].Acetate along  with propionate consumption  is feasible by  IECs, nevertheless this  possesses lesser  effectiveness in contrast to butyrate. However acetate is further correlated with induction of extra anti-inflammatory reactions   in the  gut epithelium  for instance  escalating the expression of the   tight junctional proteins  along  with anti-inflammatory cytokines     generation[123]. Apparently this gets modulated   by toll like receptor  (TLRs) , G- protein coupled receptor(GPCR) , inclusive of  FFA2 pathways stimulation,the way definition given with use of knockout(KO) mouse models.The acetate  ligand crosstalk can result  in activation  of  nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3(NLRP3) inflammasome[124] which further   is  correlated with efficacious clearance from the gut[125] as well as escalated generation  of sIgA(dendritic  cell-B cell activation ).Of the other metabolites described earlier for instance   Vitamin B  family,it has been   accepted  that these further  result  in direct modulation of  host immune reaction inclusive of theepithelial barrier (-see Yoshi,etal.[126 ],for a greater extensive reviewof this). Furthermore, it is pointed that bacteriocins might be having a double part, apart from provision of direct antimicrobial activity along with facilitating immunomodulatory activity, nevertheless, apparently this takes place more commonly with innate immune cells  like  macrophages [112]. Additionally,aromatic  lactic acid might be of significance in early life in view of their  capacity  of modulating immune function through both aryl hydrocarbon receptor[AhR]  along  with hydroxycarboxylic acid receptor3(HCA3)[64,127]. In the past 10 yrs  tryptophan  catabolites have been illustrated to be plausible crucial AhR  ligands possessing a part in controlling  intestinal   barrier along  with immune function[128]. Recently it was  illustrated that faecal quantities of  indoleacetic acid had a positive  association with  the faecal samples capacity  of  activating AhR in an AhR  reporter cell  line [88], pointing that indoleacetic acid  is a crucial AhR  ligand in early life . Nevertheless, it. Has to be estimated if indoleacetic acid along with/or microbiota obtained AhR ligands actually possess a part in intestinal   barrier production with regard to      early life.What is of significance is to have understanding that maximum work in this field has been performed on  line of adult models.  Earlier work  has pointed that microbes( inclusive of Bifidobacterium )guide  reactions  in  the neonates vs latter time points in   life[129], thereby a robust  need  exists for  detecting the manner by which microbial metabolites for instance aromatic  lactic acid along  withSCFA modulate host intestinal  reactions.

Microbial Metabolites Along With Intestinal   Motility

Frequency of stools along  with its consistency  differ with considerable   significance  as per early life   nutrition amongst along  with within at the time of the first year of early life [130],although there is minimum information  present with regard to diet , generating GM as well as persistence of   alteration of bowel habits in early life.Different microbial metabolites which have been determined in  the infant’s gut, might probably be playing a part in influencing  gut motility in early life, despite  this has not been  evaluated till now. For instance tryptamine has been observed to cause augmentation of transit   in vivo via 5 hydroxy tryptamine receptors (5-HT 4R), a receptor which is having    distinct expression in colonic epithelium [82,131].

Other tryptophan obtained catabolites might be impacting the intestinal   motility through AhR signalling in enteric neurons in the distal part of GIT[132]. Moreover, SCFA possess the  capacity  of stimulating  liberation of  serotonin((5-HT)in enterochromaffin   cells  by activation of GPCR- for instance FFA3(G protein-coupled receptor [GPR41],or FFA2(GPR43) as well as HCA2(GPR 109A ). Serotonin possesses numerous paracrine in addition to endocrine parts inclusive of facilitation   of peristalsis through the enteric nervous system (ENS)[ 133]. Secondary BAs  in addition to SCFAs might further work via TGR5 as well as FFA2/ FFA3 respectively stimulating  liberation of glucagon like peptide 1 (GLP1), peptide-YY (PYY) in enteroendocrine hormones cells as well as thus   influence colonic  motility[ 134].

 Differences in intestinal transit time have been correlated with the gut microbiome constitution along with actions in adults [ 135]. Additionally,   acknowledged  that there is presence of   great variations as well as alterations in SCFAs , secondary BAs, proteolytic metabolites at the time of first year of life ,it is probable that the gut microbiome  aids in inter along  with  intra subject differences in bowel habits at the time of  infancy. These differences might probably be correlated with the frequent gastrointestinal tract (GIT) aberrations observed in infants for instance colic along with functional constipation [ 136].  

Microbial Metabolites Along With Total Body Metabolism

SCFAs have been posited to influence total body metabolism along with inflammation following absorption as well as circulation in blood stream too far off    organs through crosstalk with host receptors [169]. Moreover, SCFA possess the  capacity  of  indirectly   influencing total body metabolism stimulating  liberation of gut hormones (for instance GLP1 along  with PYY) in enteroendocrine cells,influencing appetite as well as via  stimulation  of immune cells which control total body metabolism [79].The greater gut luminal quantities of  SCFAs, lactate in addition to succinate determined at the time of  infancy might probably be  significant for enrolment of  macrophages as well as neutrophils,working  via FFA2, HCA1 along  with GPR 91 respectively [121]. Nevertheless, in reference toearly life minimum knowledge is probably with regard to     part of SCFAs, controlling   total body metabolism along with inflammation. Greater faecal  quantities of formate at the time of  early infancy(3-4mths of age) have been correlated with a lesser    BMI z score, while greater faecal  quantities of  butyrate have been correlated with a greaterBMI z score   at 3yrs of age in case of Canadian infants[ 137]. Dependent on these findings it has been posited that the generation of the infants gut microbiome from greater formate(portraying a   lesser variation microbiome) to greater butyrate(portraying a   greater variation microbiome) might probably be playing a part in growth  direction[ 137]. Nevertheless, timing of the gut microbiome generation in infant might be significant in view of one more study conducted in Danish infants having normal weight accrual at 9mths of age possessed significantly greater faecal quantities of butyrate in contrast to infants having   greater weight accrual [77]. In agreement with this only formula feeding or early initiation complementary/solid feeding (vs at later time) at the time of infancy has been correlated with changed GM in addition to escalated chances of getting overweight in early childhood [138].

Prospective studies concentrating over the infants gut microbiome   generation are mandatory   through generation of the metabolites is actuallycausally responsible for the programming of    host metabolism, that might be stimulating long term physiological   actions which lead to facilitation or avoidance of disease.

Conclusions

In toto a proper requirement is there for untangling along with assessment of microbial metabolites as well as their crosstalk with gut microbes in addition to host cells in early life. These microbial crosstalk might be key with regard to generation of the gut microbiome as well as proper interactions amongst gut commensals in addition to host cells with short along with long term physiological sequelae . Presently maximum work regarding microbial metabolites along with ecosystem structuring have been performed in reference to adult gut. Additionally, paucity of work  with regard to crossfeeding in  the infant gut for instance Bifidobacterium  regulated HMOs metabolism along  with assessment  of formationof  SCFAs have been performed . Thereby ,need of the hour isconducting  prospective infant cohort studies subsequent  to propagation in  the diet paralleled  with gut microbiome constitution  along  with actions are the requirements with regard to advancements  in this area.Collection in addition to evaluationof longitudinal faecal samples by untargeted  metabolomics provision with regard to understanding about acknowledged  as well as innovative microbial obtained metabolites along  with their dynamics across in addition to within infants at the time of  early life is feasible  as suggestedby  Laursenet al[64].In combination with  high resolution metabolomics profiling which aids  in classification at the level of strain[12] as well as immune cell  profiling[127]would take this fieldto considerable heights. Moreover,it is warranted to form germane animal  models as well as humanorganioids simulating  early life conditions  for unravelling the functional along  with modes which are implicated in  the manner  by which microbial metabolites influence hostgeneration as well as particular pathways. This kind of understanding  would ensure generation of targeted  approaches which aid  in  healthy along  with generation in infants,that might be inclusive of modulating manipulation of    infant gut microbiotaby  utilization of probiotics prebiotics or postbiotics.Recent studies by  Holst et al.[139], illustrated that infant  formula   on supplementation  with five human milk oligosaccharides(5HMO-mix) composed of 20-fucosyllactose, 3-fucosyllactose, lacto-N-tetraose, 30-sialyllactose as well as  switches the faecal microbiome  of formula  fed infants closer to breast fed infants for instance enrichment of  Bifidobacterium as early  as 1 week which  lasted as long as 16 weeks ,thus mothers who somehow might not be able to carry out breast feeding in view of  say pregnancy induced hypertension,preeclampsia, etc this might prove to be a good substitute[139].

Bakshi etal.[140], further corroborated the concept of supplementation of Infantformula  awa  reviewed the potential pathways influenced by GM as well as the manner by which they are  correlated   with long  term actions  on health(see Figure 5).

Figure 5: Courtesy ref no-140-Potential pathway entailing gut microbiota and its link to long-metabolic health. HMO, Human milk oligosaccharides; SCFAs, Short-chain fatty acids; PYY, Peptide YY; IGF-1, Insulin-like factor-1; miRNA, non-coding microRNA; GLP-1, Glucagon-like peptide-1.

Figure 6: Courtesy ref no-141-Supposed effects on the intestinal barrier, immunity, and microbiota of infant formula supplemented with pre-, pro-, syn-, and postbiotics compared to breastfeeding. Legend: BMOs—bovine-milk-derived oligosaccharides; FOSs—fructo-oligosaccharides; GOSs—galacto-oligosaccharides; HMOs—human milk oligosaccharides.Yelverton etal.[142], performed  a secondary evaluation  of healthy pregnant  women with regard to Microbe Mom   study;a double blind,  randomized   controlled  trial  where maternal probiotics supplementation(Bifidobacterium breve 702258) vs placebo in antenatal along  with uptill 3mths postpartum women(n=118) with a min of 1estimate of well being over mean 33yrs(SD3.93)agewise along  with uptill 25.09kg/m2BMI.Sole breast feeding  was started by 65%(n=74). Continuation of any breast feeding   by 69%(n=81) subsequent  to 1 mth was done.In early as well as  latter part ofpregnancy87%(n=97/111) in addition to  94%(n=107/114) possessed > well being scores. Well being  was not correlated with  infant microbial  variation at  1mth.Inadjusted evaluation, sole breast feeding at the time of   discharge was correlated with  infant  microbial  beta variation(PC;0.254,95%CI  0.006,0.038).At   1mth postpartum, any breast feeding correlated with  infantmicrobial  alpha variation(Shannon index;-0.241,95%CI-0.49,-0.060),along  with spp found ;(-0.325, 95%CI  -0.307,-0.060)  as well as infantmicrobial  beta variation(PC2;0.319,95%CI 0.013,0.045). Sole breast feeding at   1mth postpartum was correlated with  infantmicrobial  alpha variation(Shannon index;-0.364,95%CI-0.573,-0.194), in addition to     Simpson index;0.339,95%CI0.027,0.091), along  with infants spp found ;(-0.271, 95%CI  -0.1712,-0.037).Thus Conclusionsdrawn were  breast feeding practices were  correlated with lesser  microbial  variations in infants at  1mth postpartum that is plausibly of advantages  in aiding  in enrichment of  Bifidobacterium.

Recently Juge N[[143],  briefed about  how  Ruminococcus gnavusa  is a human gut symbiont, part of the infant as well as adult gut microbiota along with  correlated with intestinal in addition to   extra-intestinal conditions. Mechanistic part of how R. gnavus  aids in adaptation to the gut are strain- particular as well as  reinforced  by thecapability of R. gnavus strains to use mucin as well as dietary glycans a along  with generate bacteriocins in addition toadhesins. Different plausible mediators  strengthening  the correlation  amongst   R. gnavus strains along  with  diseases have been isolated inclusive of the capability of evoking a pro- or anti-inflammatory conditions   host reactions  in addition to  modulation  of host metabolism, secondary bile acids along  with tryptophan   metabolic pathways.Dependent on escalating  corroboration    from metagenomics studies in humans as well as  functional evaluation in vitro as well as  in mouse models, R. gnavus is being illustrated in  the form of  a major actor in impacting  health along with  diseaseresults  from  infants to the  older adults[143& rev in detail in 144](see Figure7).

Figure 7: Courtesy ref no-143-Graphic Abstract.

References

  1. Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong MC, et al. Temporal development of the Gut Microbiome in early childhood from the TEDDY study. Nature. 2018; 562: 583-588.
  2. Dominguez-Bello MG, Godoy-Vitorino F, Knight R, Blaser MJ. Role of the microbiome in Human Gut. 2019; 68: 1108-1114.
  3. Ratsika A, Codagnone MC, O’Mahony S, Stanton C, Cryan JF. Priming for early life nutrition and microbiota- gut- brain axis. 2021; 13: 423.
  4. Turroni F, Milani C, Duranti S, LugliGA, Bernasconi S, Margolles A, et al. The infant Gut Microbiome as a microbial  organ influencing host well-being. Ital J Paediatr. 2020; 46: 16.    
  5. Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamicsand stabilization  of the human Gut microbiome during the First year of  Cell host Microbes. 2015; 17: 690-703.
  6. Zmora N, Suez J, Elinav E. You are what you eat:diet, health and gut microbiota. Nat Rev Gastroenterol Hepatol. 2019; 16: 35-56.
  7. Cowan CSM, Dinan TG, Cryan JF. Annual research review: critical windows-the microbiota- gut- brain axisin neuro J Child Psychol Psychiatr. 2020; 61: 353-371.
  8. Swanson KS, Gibson GR, Hutkins R, Reimer RA, Reid G, Verbeke K, et al. The International Scientific Association of Prebiotics and Probiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. 2020; 17: 687-701.
  9. Zolkiewicz, J,Marzec A, Ruszczynski M, Feleszko W. Posbiotics-a step beyond Pre and Probiotics. 2020; 12: 1-17.
  10. Murphy K, Ross RP, Ryan CA, Dempsey EM, Santon CP. Prebiotics and Probioticsfor the prevention of necrotizing enterocolitis. Front Nutr. 2021; 8: 1012087.
  11. Quigley EMM, Gajula P. Recent advances in modulating the F1000res. 2020; 9: 46.
  12. Enav H, Backhed F, Ley RF. The developing infant Gut microbiome: A strain level Cell host Microbes. 2022; 30: 627-638.
  13. Roager HM, Dragsted LO. Diet derived microbial metabolites in health and disease. Nutr Bull 2019; 44: 216-227.
  14. Berry SE, Ana MV, David AD, Asnicar F, Mazidi M, Haya AH, et al. Human postprandial responses to potential to food and precision nutrition. 2020; 26: 1-10.
  15. Kulvinder KK, Allahbadia GN, Singh M. An update on the Generation of the Gastrointestinal Tract in Neonates with Specific Emphasis on the Hurdles Encountered in Feeding Preterm Infants with Associated Complications like Gut, Brain &G-B Axis  Generation: A Narrative Review. Journal of BioMed Research and Reports. 2022; 1: 1-15.
  16. Kulvinder KK, Allahbadia GN, Singh M. Possible protective impact of Breast Feeding on future Reprogramming&Colonization of Neonates through Sirtuins-A MiniReview.  Accepted for Publication in SCPaediatrics Journal. 2023.
  17. Kulvinder KK, Allahbadia GN, Singh M. An update on the Association of Gut-Liver Axis with Gut Microbiome Dysbiosis Correlated NAFLD along with NAFLD- HCC with Potential Therapeutic Approaches: a systematic review. Accepted for Publication in. J Hepatol. 2022.
  18. Munyaka PM, Khafipour E, Ghia JE. External influence of early childhood establishment of gut microbiota and subsequent health Front Paediatr. 2014; 2: 109.
  19. Dominguez-Bello MG, Costello EK, Contreas M, Megris M, Hidalgo G, Fierer N, Knight R. Delivery modes shape the acquisition and the structure of initial microbiota across multiple body habitats in the new born. Proc NatlAcad Sci. 2010; 107: 11971-5.
  20. Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, et al. Stunted microbiota and opportunistic pathogen colonization  in caesarean section Nature. 2019; 574: 117-121.
  21. Laursen MF. Gut microbiota development: Influence of diet from infancy to toddlerhood. Ann Nutr Metab. 2021; 77: 21-34.
  22. Hills CJ, Lynch DB, Murphy K, Ulaszewska M, Jeffrey IB, O’SheaCA, etal. Erratum to: evolution of gut microbiota composition from birth to24 weeks in theinfantmet Microbiome. 2017; 5: 21.
  23. Reyman MA, Van Houten MA, Watson RI, Chu MLJN, Arp K, Waal DWJ, et al. Effects of early life  antibiotics  on  the developing infant Gut microbiome and resistome:a randomized trial. Nat Commune.2022; 13: 893.
  24. Hernandez E, Bargiela R, Diez MS, Friedrich AS, Perez-Cobal AE, Gosalbes MJ, et al. Functional consequences of microbial shifts in the human Gastrointestinal Tract linked to antibiotic treatment and obesity. Gut Microbes. 2013; 4: 306-315.
  25. Murphy K, Curley D, O’Callaghan TF, O’Shea CA, Dempsey EM, O’Toole PW, et al.The composition of human and infants faecal microbiota over the First 3 mths of life: a pilot study. Sci Rep. 2017; 7: 40597.
  26. Laursen MF, Zachariassen G, Bahl MI, Bergstrom A, Host A, Michaelsen KF, etal. Having older siblingsis associated with gut microbiota development during early childhood. BMC Microbiol. 2015; 15: 154.
  27. Victoria CG, Bahl R, Barros AGD, Franca GVA, Horton S, Krasevec J, et al. Breast feeding in the 21st century: epidemiology, mechanisms and lifelong   Lancet. 2016; 387: 475-490.
  28. Ahern GJ, Hennessy AA, Anthony RC, Paul RR, Stanton C. Advances in infant formula Annu Rev Food Sci Technol. 2019; 10: 75-102.
  29. Davis JCC, Totten SM, Huang JO, Nagshbadi S, Kirmiz N, Garrido DA, et al. Identification of oligosaccharides in faeces of breast fed infants and their correlation with gut microbial Mol Cell Proteomics 2016; 15: 2987-3002.  
  30. Chung S, Bode L, Hall DA. Point of care human milk testing for maternal secretor status. Anal Bioanal Chem. 2022; 414: 3187-3196.
  31. Cabrera-Rubio R, Kunz C, Rudloff S, Garcia-Mantrana I, Crehua-Gaudiza E, Martinez-C C, et al. Association of maternal secretor status and human milk oligosaccharides with milk microbiota: an observational pilot study. J Paediatr Gastroenterol Nutr. 2019; 68: 256-263.
  32. Kumar H, Wacklin P, Nakphaichit M, Loyttyniemi E, Chowdhari S, Shouche Y, et al. secretor status is strongly associated with microbial alterationsobserved during 2015; 10: e1034623.
  33. Lodge CJ, Lowe AJ, Milanzi E, Bowatte G, Abramson MJ, Tsimiklis H, et al. Human milk oligosaccharides profiles and diseases upto 18 years. J Allergy Clin Immunol. 2021; 147: 1041-1048.
  34. Parnanen KMM, Hutmao J, Maarkanen M, Satokari R, Kelleher SL, Lamendella R, et al. Early life formula feeding is associated with gut microbiota alterations and increased antibiotics res load. Am J Clin  2022; 115: 407-421.
  35. Rehman SF, Olm MR, Morowitz MJ, Banfield IF, Segata N. Machine learning leveraging genomes from metagenomes identifies influential antibiotics resistance genes in the infant gut microbiome. mSystems  2018; 3: e00123-11.
  36. Sakanaka M, Gotoh A, Yoshida K, Odamaki T, Koguchi H, Xiao JZ, etal. Varied pathways of infant gut associated Bifidobacterium to assimilate human milk oligosaccharides: Prevalence of the gene set and its correlation with Bifidobactera rich microbiota Nutrients 2019; 12: 71H.
  37. He X, Parenti M, Grip T, Domellof M, Lonnerdal B, et al. Faecal microbiome and metabolome fed bovine MFGM supplemented formula or Standard formula with breast fed infantsas reference:a randomized controlled   Sci Rep. 2019; 9: 11589.
  38. Wang Z. Klipfell E, Bennett BJ, Koeth R, Levison BS, DuGar B, et al. gut flora metabolism of phosphatidyl choline promotes cardiovascular disease. Nature. 2011; 472: 57-63.
  39. Koh A, Molinaro A, Khan M, Schmidt, Manneras HolmL, et al. Microbially produced imidazole propionate impairs insulin signalling through mTORC1 2018; 175: 947-961.
  40. Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, et al. A cardiovascular disease linked gut microbial metabolites act via adrenergic 2020; 180: 862-877.
  41. Wu I, Hsu K, Lee CC, Sun CY, Hsu HJ, Tsai CJ, et al. p-cresyl sulphate and indoxyl sulphate predict progression of Chronic Kidney Disease. Nephrol Dial 2011; 26: 938-947.
  42. Bosheva M, Tokodi I, Krasnow A, Pederson HK, Lukjancenko O, Eklund AC, et al. Infant formula with a specific blend of five human milk oligosaccharides drives the gut microbiota development and improves gut maturation markers: a randomized controlled Front Nutr 2022; 9: 920362.
  43. Laursen MF, Bahl MI, Michaelsen KF, Licht TR. First foods and gut microbes. Front Microbiol. 2017; 8: 356.
  44. Leong C, Haszard JJ, Lawley B, Otal A, Taylor RW, Szymlek Gay EA, et al. Mediation analysis as a means of identifying dietary components that differentially affect  the faecal microbiota of the infants weaned by modified baby -led and traditional approaches. Appl Environ Microbiol. 2018; 84: e00914-18.  
  45. Laursen MF, Michaelsen KF, Andersen LBB, Molgaard C, Trolle E, Bahl MI, Licht TR. Infant gut microbiota development is  by transition to family foods independent  of maternal mSphere 2016; 1: 1-16.  
  46. Roswall J, Olsson LM, Kovatcheva-Datchary P, Nilsson S, Tremaroli V, Simon MC, et al. developmental trajectory of the healthy human gut microbiota during the First 5years of Cell Host Microbes   2021; 2930: 765-776.
  47. Subramaniam S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam M, et al. Persistent gut microbiota immaturity in malnourished Bangladesh children . 2014; 510: 417-421
  48. Blanton LV, Charbonneau MR, Salih T, Barratt MJ, Venkatesh S, Ilkaveya O, et al. Gut bacteria that prevent growth impairment transmitted by microbiota from malnourished Science. 2016; 351; 10.
  49. Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The human microbiome and child growth- First 1000days and beyond. Trends Microbiol. 2019; 27: 131-147.
  50. Chen RY, Mostafa I, Hibberd MC, Das S, Mahfuz M, Naina NN, et al. A microbiota directed food intervention for undernourished N Engl J Med. 2021; 384: 1517-1528.
  51. Barratt MJ, Nuzhat S, Arzamasov K, Frese SA, Arzamasov AA, Sarker SA, et al. Bifidobacterium infantis treatment promote weight gain in Bangladeshi infants with severe acute Sci Transl Med. 2022; 14: eabk1107.
  52. United Nations Children ‘sFund(UNICEF), World Health Organization& International Bank for Reconstruction and development /The World Bank. Levels and trends in Child :key findings of the 2021 edition of the Joint Child Malnutrition estimates. 2021
  53. Grote V, Verduci E, Scaglioni S, Vecchi F, Contarini G, Giovannini M, et al. Breast milk composition and infants nutrient intakes during First 12 mths of  life . Eur J  Clin  2016; 70: 250-256.
  54. Baumann-Dudenhoeffer AM, D’Souza AW, Tarr PI, Warner BB, Dantas G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbes. NatMed. 2018: 24: 1822-9.
  55. Roager HM, Stanton C, Hall Microbial metabolites as modulators of the infant gut microbiome andhost microbial interactions in early life. Gut Microbes. 2023; 15: 2192151.
  56. Koh A, Vaader DF, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short chain fatty acids as key bacteria metabolites. 2016; 165: 1332-45.
  57. McNeil NI. The contribution of thelarge intestine to the energy supplies in man. Am J Clin Nutr 1984; 39: 338-342.
  58. Vaader DF, Kovatcheva-Datchary P, Vinera J, Zitoun C, Duchampt A, Backhed F, et al. Microbiota generated metabolites  promote metabolic  benefits  via  gut- brain neural 2014; 156: 84-96.
  59. denBesten G, Lange K, Havinga R, van DTH, Gerding A, vanEunen K, et al. Gut derived short chain fatty acids are vividly assimilated into host carbohydrate and   Am J Physiol Gastroenterol Liver Physiol. 2013; 305: G900-G5910.
  60. Tsukuda N, Yahaki K, Hara T, Watanabe Y, Matsumoto H, Mori H, et al. Key bacterialtaxa and metabolic pathways affecting  gut short chain fatty acids profiles in early life.Isme J. 2021; 15: 1517-1528.
  61. Sillner N, Walker A, Lucio M, Maier TV, Bazanella M, Rychlik M, et al. longitudinal profiles of dietary and microbial metabolitesin formulas and breast fed infant Front Mol Biosci. 2021; 8: 660456.
  62. Duncan SH, Louis P, Flint HJ. Lactate utilizing ed bacteria isolated from human faeces that produce butyrate as a major fermentation product. Appl Environ Microbiol. 2004; 70: 5810-5817.
  63. Bridgman SI, Azad MB, Field CJ, Haqq AM, Becker AB, Mandhane PJ, et al. Faecal short chain fatty acids variations by breast feeding status in infants at 4months: differences in relative versus absolute Front Nutr. 2017; 4.
  64. Laursen MF, Sakanaka M, vonBurg N, Morbe U, Andersen D, Jm M, et al. Bifidobacterium species associated with breast feeding produce aromatic lactic acids in the gut . Nat Microbiol. 2021; 6: 1367-1382.
  65. Thompson P, Medina DA, Garrido D. Human milk oligosaccharides and infants gut Bifidobacteria: molecular strategies for their utilization. Food 2018; 5: 37-46.
  66. Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshmura K, et al. Bifidobacteria can protect from enteropathogenic infection through protection of acetate. 2011; 469: 543-547.
  67. Soyilmaz B, Miks MH, Rohrig CH, Matwiejuk M, Meszaros-Matwiejuk A, Vigsnaes LK. The mean of milk:a review of Human milk oligosaccharides Concentrations throughout lactation. Nutrients. 2021; 13: 2737.
  68. Differding MK, Benjamin-Neelon SE, Hoyo C, Ostbye T, Mueller NT. Timing of  complementary feeding is associated with and gut microbiota diversity and composition and short chain fatty acids Concentrations over the First few years of  BMC Microbiol. 2020; 20: 56.
  69. Michelsen KF, Grummer Strawn L, Begin F. Emerging issues in complementary feeding; global aspects. Maternal -Child Nutr. 2017; 13: e12444.
  70. Singh R, Chandrashekharappa S, Bodduluri SR, Baby BV, Hegde B, Kotla NG, et al. Enhancement of the gut barrierintegrity by a microbial metabolite through Nrf2 Nat Commun. 2019; 10: 89.
  71. Chen H, Pk N, Yang Y, Ce R, Aa B, Kuchroo M, et al. Aforward genetic  screening reveals gut microbiota metabolites that modulationhost physiology. Cell 2019; 177: 1217-1231.
  72. Brink LR, Mercer KE, Piccolo BD, Chintapalli SV, Elolimy A, Bowlin AK, et al. neonatal diets alter faecal microbiota and the metabolomic profiles at different ages in infants fed breast milk or formula. Am J Clin Nutr. 2020; 111: 1190-1202.
  73. Newgard CB, An J, Bain R, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched chain amino acidsrelated metabolic signatures that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab 2009; 9: 311-326.
  74. Kok CR, Brabec B, Chichlowski M, Harris CL, Moore N, Wampler JL, et al. Stool microbiome,pH and short/ branched chain fatty acids in infants receiving extensively hydrolyzed  formula, amino acids formula  or human milk through two months of BMC Microbiol. 2020; 20: 337.
  75. Agostoni C, Boniglia E, Carrato B, Riva C, Sanzini E. Free amino acids content in infant standard formulas: comparison with human milk. J Am Coll Nutr. 2000; 19: 434-438.
  76. He X, Sotelo-Orozco J, Rudolph C, Lonnerdal B, Slupsky CM. The role of proteins and free amino acids on intake, metabolism, and gut microbiome: a comparison formulas breast fed and formula fed rhesus monkey infants. Front Paediatr. 2020; 7: 563.
  77. Pekmez CT, Larsson MW, LindO MV, Vasquez Manjarrez MN, Yonemitsu C, Larnkjaer A, et al. Breast milk lipids and oligosaccharides influence branched short chain fatty acids concentrations in infants with excessive weight Mol Nutr Food Res. 2020; 64: 1900977.
  78. Suarez L, Moreno–Luque M, Martinez-A I, Gonzalez N, Campo P, Huerta-Cima P, et al. Amine variations in Faecal content in the first weeks of life of newborns in relation to breast feeding  or  infant Br J Nutr. 2019; 122: 1130-1141.
  79. Ma,LJZ, Zhang W, Zhang C, Zhang Y, Mei H, et Comparison of gut microbiota inexclusively Breast fed and formula fed babies:a study of 91 term infants. Sci Rep. 2020; 10: 15792.
  80. Pugin B, Barcik W, Westermann P, Heider A, W A, Helling P, et al. A wide diversity of from the human gut produces and degrades biogenic MicrobEcolHealth Dis. 2017; 28: 1353881.
  81. Sagheddu V, Patrone V, Miragoli F, Puglisi E, Morelli L. Infant early gut colonization by Lachnospiraceae: high frequency of Ruminococcus gnavus. Front 2016; 4: 57.
  82. Williams BB,Van Benschoten A,Cimermanic P,D M, Zimmermann M, Taketani M, etal. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell host Microbes   2016; 16: 495-503.
  83. Nejrup RG, Licht TR, HellgrenLI. Fatty acid composition and phospholipid types used in infant formulas modifies the establishment of human gut bacteria in germ free mice. Sci Rep. 2017; 7: 1-11.
  84. Hennessy AA, Barrett E, Paul Ross R, Fitzgerald GF, Devery R, Stanton C. The production of  conjugated α-linolenic  acid , γLinolenic Acid,Stearidonic Acids by strains of Bifidobacteria and  propiono bacteria. Lipids 2012; 47: 313-327.
  85. Mei Y, Chen H, Yang B, Zhao J, Zhang H, Chen W. Research progress on conjugated linolenic acid bioconversion in Bifid bacterium. Int J Food Microbiol. 2022; 369: 109593.
  86. Kim JH, KimY, Kim YJ, Park Y. Conjugated linolenic acid: potential health benefits as a functional food ingredient. Annu Rev Food Sci Technol.  2016; 7: 221-244.
  87. Bevan SR, Stamnes KA, Lin DL, Panzer AR, Fukui E, McCauley K, etal. Elevated faecal 12,13-diHOME Concentrations in neonates at high risk for asthma is producedby gut bacteria and   impedes immune tolerance . Nat Microbiol 2019; 4:1851-1861.
  88. Martin FP, Tytgat HLP, Krogh Pedersen H, Moine D, Eklund AC, Berger B, etal. Host microbial cometabolites modulated by Human milk oligosaccharides relate to reduced risk for respiratory tract infections. Front Nutr. 2022; 9:1752.
  89. Wahlstrom A, Sayin SI,Marschall HU, Backhed F. Intestinal crosstalk between bile acids and microbiotaand its impact on host metabolism. Cell Metab. 2016; 24: 41-50.
  90. Ridlon JM, Kang DJ, Hylemon PB. Bile salts biotransformations by human intestinal bacteria. JLipids Res   2006; 47: 241- 259.
  91. Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory JL ipids Res. 2009; 50: 1509-1520.
  92. Tanaka H, Sanefulji M, Morokuma S, Yoden M, Momoda R, Sonomoto K, et al. The association between gut microbiota development and maturation of intestinal bile acids metabolism in the First 3years of healthy Japanese infants. Gut Microbes. 2020; 11: 2530-235.
  93. Tanaka H, Doesburg K, Iwasaki T, MierauI. Screening of lactic acid bacteria for Bile salts Hydrolase activity. J Dairy Sci. 1999; 82: 2530-2535.
  94. Xiong JJ, Hu HH, Xu CZ, Yin JW, Liu M, Zhang LZ, et al. Developmental patterns of faecal bile acids in healthy neonates and children. Med Sci Monit. 2021; 27: e928214.
  95. vanBest N, Rolle Kampczy kU, Schaap FG, Basic M, Olde Damiuk SWM, Bleich A, etal. Bile acids drive the newborn’s gut microbiome maturation. Nat Commun.  2020; 11: 3692.
  96. Schwab C, Ruscheweyh HJ, Bunesova V, Pham V, Beerenwinkel N, Lacrol C. Trophic  interactions of  infant  Bifidobacterium and  eubacterium hallii during  L-fucose  and  fucosyllactose degradation. Front Microbiol. 2017; 8: 95.
  97. Lawson MAE, O’NeilI J, Kujawska M, Gowrinath JavvadiS ,Wijeyseekera A, Flegg Z,etal. Breast milk derived Human milk oligosaccharides promote Bifidobacterium interactions within a single ecosystem. Isme J. 2020; 14: 635-648.
  98. Masi AC, Stewart CJ .Untangling Human breast milk oligosaccharides and infant gut microbiome . I Science. 2022; 25: 103542.
  99. Kijner S, CherA,Yassour M. The infant gut commensal Bacteroides dorei presents a generalized transcriptional response to various Human milk oligosaccharides. Front Cell Infect Microbiol. 2022; 12: 285.
  100. Sela DA, Mills DA. Nursing our microbiota: molecular linkages between Bifidobacterium and milk oligosaccharides. Trends Microbiol. 2010; 18: 298-307.
  101. Fernandez- Julia P, Commane DM, Van Sinderen D, Munoz- Munoz J. Cross feeding interactions between human gut commensals belonging to the Bacteroides and Bifidobacterium genera when grown on Dietary glycans. Microbiome Res Rep. 2022; 1:12.
  102. Cheng CC, Duar RM, Lin X, Perez Munoz ME, Tollenaar S, Oh JM, et al. Ecological importance of Cross feeding of the intermediate metabolite 1,2, propanediol between bacterial gut  symbionts . Appl Environ Microbiol.  2020; 26: e00190-220.
  103. Walsh C, Lane JA, Van Sinderen D, Hickey RM. Human milk oligosaccharides -snaring by a consortium of infant derived Bifidobacterium species. Sci Rep.  2022; 12: 1-14.
  104. Ojima MN, Jiang L, Aa A, Yoshida K, Odamaki T, Xiao J, etal. Priority effects shape the structure of infant type Bifidobacterium communities on Human milk oligosaccharides. Isme J. 2022; 14:1-15.
  105. Alcon Giner C, Dalby MJ, Caim S, Ketskemety J, Shaw A, SimK, et al. Microbiota supplementation with Bifidobacterium and Lactobacillus modifies the  preterm    infants gut microbiota and metabolome: an observational study. Cell Rep Med.   2020; 1:100077.
  106. Zhang S, Dogan B, Guo C, Herlekar D, Stewart K, Scherl EJ, et al. Short chain fatty acids modulate the growth and virulenceof pathosymbiont Escherichia Coli and host response. Antibiot (Basel).  2020; 9: 1-20.
  107. ChiaLW, Mank M, Blijenberg B, Bonders RS, VanLimpt K, Woperies H, et al. Crossfeeding between Bifidobacterium infantis andAnaerostipes caccae on lactose and Human milk oligosaccharides. Benef Microbes 2021; 12: 69-83.
  108. Glover JS, Ticer TD, Engevik MA. Characterizing the mucin degrading capacity of the human gut microbiota. Sci Rep. 2022; 12: 8456.
  109. Garber JM, Hennet T, Szymanski CM. Significance of fucose in intestinal health and disease. Mol Microbiol2021; 115:1086-1093.
  110. Heilbronner S, Krismer B, Brotz Oesterhelt H, Peschel A. The microbiome shaping roles of bacteriocins. Nat Rev Microbiol. 2021; 19: 726-739.
  111. Martinez FAC, Balciunas EM, Converti A, Cotter PD, deSouzaOlievieria RP. Bacteriocin production by Bifidobacterium species. A review. Biotechnol Adv. 2013; 31: 482-488.
  112. Javvadi S, Kujawska M, Paap D, Am G, Jordan A, Ma L, et al. A novelbacteriocinproduced by Bifidobacterium longum subspp infantis has dual anti-microbial and immunomodulatory activity. bioRxiv. 2022; 2022:
  113. Zafar H, Safar MH. Gut Bacteroides species in health and disease. Gut Microbes. 2021; 13: 1-20.
  114. Oliphant K, Allen-Vercoe E. Macro nutrients metabolism by the human gut microbiome: major fermentation by products and their impact on host health. Microbiome. 2019; 7: 1-15.
  115. Lin R, Liu W, Piao M, Zhu H. A review of the relationship between the gut microbiota and amino acids metabolism. Amino Acids. 2017; 49: 2083-2090.
  116. Degnan PH, Taga ME, Goodman AL. Vitamin B12 as a modulator of gut microbial Ecology.  Cell Metab 2014; 20: 769-778.
  117. Wan Z, Zheng J, Zhu Z, Sang L, Zhu J, Luo S, et al. Intermediate role of gut microbiota in Vitamin B nutrition   and its influence on human health. Front Nutr. 2022; 9.
  118. Uebanso T,  Shimohata T, Mawatari K, Takahashi A. Functional roles of B - Vitamins in the gut and gut microbiome. Mol Nutr Food Res. 2020; 64: 2000426.  
  119. Vaales Colomer M, BiancoMiquez A, Manghi P, Asnicar F, Dubois L, Golzato D, et al. The person to person transmission landscape of the gut and oral microbiome. Nature. 2022; 614:125-135.
  120. Wikoff WR, Anfora AT, Liu J, Schultz PJ, Lesley SA, Peters EC, etal. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc NatlAcad Sci.  2009; 106: 369698-369703.
  121. Husted AS, Traulesen M, Rudenko O, Hjorth SA, Shwarz TW.GPCR mediated signalling of metabolites. Cell Metab 2017; 25: 777-796.
  122. Salvi PS, Cowles RA. Butyrate and the intestinal epithelium: modulation of proliferation and inflammation in homeostasis and disease. Cells. 2021; 10: 1775.
  123. Ma J, Piao X, Mahfuz S, Long S, Wang J. The interaction among gut microbes, the intestinal barrier and short chain fatty acids. Animal Nutr. 2022; 9: 159-174.
  124. Macia L, Tan J, oVieira AT, Leach K, Stanley D, Luong S, et al. Metabolite sensing GPR43 and GPR 109A facilitate dietary fiber induced gut homeostasis through regulation of the inflammasome. Nat Commun   2015; 6: 6734.
  125. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells  to promote inflammatory response in mice. Gastroenterology 2013; 145: 396-406.
  126. Yoshi K, Hosomi K,SawaneK, Kunisawa J .Metabolism of  dietary and microbial Vitamin B  family in  the regulation of host immunity. Front Nutr. 2019; 6: 48. 
  127. Henrick BM, Rodriques L, Lakshmikanth T, Pou C, Henckel E,Arzoomand A, etal. Bifidobacteria mediated immune system imprinting early in life. The shit. Cell. 2021; 184: 3884-3898.
  128. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018; 9: 3294.
  129. Kiu R, Treveil A, Harnisch LC, Caim S, Leclaire C, Van Sinderen D, et al. Bifidobacterium breve UCC22003 induces a distinct global transcriptomic program in neonatal murine intestinal epithelial cells. I Science 2020; 23:101336.
  130. Moretti E, Rakza T, Mestdagh B, Labreuche J, Turck D. The bowel movement characteristics of tio exclusively breast fed and    exclusively formula fed infants differ during the First 3 months of early life. Acta Paediatrica Int J Paediatrics. 2019; 108: 877-881.
  131. Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR,T L,Grover M,etal. Gut microbiota produce tryptamine activates an epithelial G- protein coupled receptor to increase colonic secretion.  Cell host Microbes.   2018; 23: 775-785.e5
  132. ObataY, Castano A,B S, BonFrauches AC ,Fung C ,Fallesten T, et al. Neuronal programming by microbiota regulates intestinal physiology. Nature. 2020; 578: 284-289.
  133. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, etal. Short chain fatty acids regulate the enteric neurons and control Gastro intestinal motility in rats. Gastroenterology. 2010; 138:1772-1782.
  134. Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Bile acids receptor and gastrointestinal functions. Liver Res. 2019; 3: 31.
  135. Prochazkova N, Faloney G, Lo D, TrL,R J, HmR, etal. Advancing human gut microbiota research by considering gut transit time. Gut. 2022; 72:180-191.
  136. Vandenplas Y, Hauser B, Salvatore S. Functional Gastrointestinal disorders in infancy: impact on the health of infant and family. Paediatr Gastroenterol Hepatol  2019; 22: 207-216.
  137. Bridgman SI, Malmathuge N, Mandal R ,Field CJ,Haqq AM, Mandhane PJ, et al. Childhood body mass index and associations with infant gut metabolites and secretory IgA: findings from a prospective cohort study. Int J Obes(Lond). 2022; 46: 1712-1719.
  138. Differding MK, Doyon M, Bouchard L, Perron P, Guerin R, Asselin C, etal. Potential interactions between timing of infant complementary feeding and breast feeding in determination of early Childhood gut microbiota composition and BMI. Paediatr Obes. 2020; 15: e12642.
  139. Holst AQ, Myers P, Rodriques Garcia P, Hermes GDA, Melsaether C, Baker A, et al. Infantformula supplemented with five human milk oligosaccharides shifts the faecal microbiome of formula fed infants closer to breast fed infants. Nutrients 2023; 15: 3087.
  140. Bakshi S, Kumar Paswan V, PrakashYadav S, KumarBhincchar B, Kharkal S, Rose H, et al. A Comprehensive review on Infantformulas: nutritional and functional constituents, recent trends in processing and its impact on Infants gut microbiota. Front Nutr 2023; 10:1194679.
  141. Lemoine A, Tounian P, Adel-Patient K, Thomas M. Pre, pro -, syn- and postbiotics in Infantformulas:what are the immune benefits for Infants? Nutrients 2023; 15: 1231.
  142. Yelverton CA, Killeen SL, Feehily C, Moore RL, Callaghan SL, Geraghty AA, etal. Maternal breast feeding is associated with off spring microbiome diversity:a secondary analysis of the Microbe Mom randomized   controlled  Front Microbiol. 2023; 14: 1154114.
  143. Juge H. Microbe profile: Ruminococcus gnavus:the yin and yang of human gut symbionts. Microbiology(Reading) 2023;169: 001383
  144. Crost EH, Coletto E, Bell A, Juge N. Ruminococcus gnavus:a friend or foe for human health. FEMS Microbiology Reviews. 2023; 47: 1-23.