Next Article in Journal
Inhibition of the Growth of Escherichia coli and Staphylococcus aureus Microorganisms in Aesthetic Orthodontic Brackets through the In Situ Synthesis of Ag, TiO2 and Ag/TiO2 Nanoparticles
Previous Article in Journal
Niacinamide Antimicrobial Efficacy and Its Mode of Action via Microbial Cell Cycle Arrest
Previous Article in Special Issue
The Relationship between Exposome and Microbiome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases

Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, 20157 Milan, Italy
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(8), 1582; https://doi.org/10.3390/microorganisms12081582
Submission received: 28 June 2024 / Revised: 29 July 2024 / Accepted: 30 July 2024 / Published: 2 August 2024
(This article belongs to the Special Issue Intestinal Dysbiosis)

Abstract

:
Probiotics are “live microorganisms which, when administered in adequate amount, confer health benefits on the host”. They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.

1. Introduction

The human microbiota plays a critical role in maintaining health and serves as a valuable resource for host defense. The early years of life are critical for the proper development of the gut microbiota, which is shaped by various factors, including the mode of feeding and delivery. The human gut contains 1014 bacteria and many other microorganisms, such as Archaea, viruses, and fungi, which are ten times more numerous than the total number of human cells. More than 95% of the gut microbiota can be classified into four major groups: Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria [1]. After bacterial colonization in infancy, the intestinal microbial composition continues to develop and reaches its adult composition by 2–3 years of age, becoming unique to everyone, with a vast difference between health and disease [2,3].
Intestinal dysbiosis, an imbalance or disruption in gut microbial populations, due to quantitative or qualitative changes in composition and/or metabolic activities and distribution [4], is correlated to a plethora of gastrointestinal (GI) pathological conditions [5], such as inflammatory bowel disease (IBD) [6], Helicobacter pylori infection [7,8], celiac disease [9], nutritional disorders [10], and many other gastrointestinal disorders [11]. Intestinal dysbiosis is also observed in allergic conditions like atopic dermatitis, allergic rhinitis, asthma [11], obesity [12], and autism [13,14]. So, the most worrying question is what would be the origin of gastrointestinal dysbiosis? Specifically, Bajinka et al. [15] reviewed extrinsic factors that influence gut microbiota with positive or negative effects and, in some cases, are associated with intestinal dysbiosis, including dietary (high fat, fiber, animal fat, amino acids, gluten, sucralose), diet therapy (Mediterranean, Western, or Vegan diet), antibiotics and drugs use, oxidative stress, and socioeconomic status [16,17]. Certainly, the use of biotics, the focus of this review, influences the composition and activity of the gut microbiota and, if prolonged or incorrect can contribute to intestinal dysbiosis.
The differences in the diversity and development of the gut microbiota in infants may also be explained by other factors. Among them, home biodiversity (family size and exposure to high levels of endotoxin in house dust) [18], the surrounding environment [19], the presence of siblings and family members [20], the environment while growing up (i.e., urban vs. rural), geographical location/population, and hygienic practices can impact microbial diversity [21]. In a recent study examining the Canadian population, it was reported that there has been a notable increase in IBD intestinal dysbiosis among children aged 0–5 years, while the incidence has remained steady in children aged 6 years and above [22]. Data from Asia and South America also suggest a similar trend, albeit with fewer patients overall [23]. The role of bacteria, fungi, and viruses in the development of IBD has been widely demonstrated and extensively reviewed elsewhere [14]. Moreover, since children are considered the best model to study the pathogenesis of IBD [15], our review aims to provide as much detail as possible in pediatric age. Figure 1 summarizes the potential factors contributing to gut dysbiotic conditions.

Development of Infant Intestinal Microbiota

Alterations in infants’ gut microbiota composition during their first year of life are influenced by various factors, and delivery mode and cessation of breastfeeding are two key factors [24,25] (Figure 2). Infants delivered by cesarean section (CS) indeed were found to have abnormal gut microbiota characterized by an increased presence of Clostridium, Clebsiella, and Enterococcus and a decreased presence of Bifidubacterium in respect to vaginally delivered ones [26,27]. One of the possible causes of these alterations in newborns is the need for antibiotic consumption by mothers undergoing cesarean section [28,29]. Moreover, mounting evidence suggests that an irregular gut microbiota composition due to delivery mode influences not only the subsequent regulation of immune response [30] but also may be linked to several clinical conditions [31]. Increased risk of obesity [32], celiac disease, type 1 diabetes [33], and asthma [34] have indeed been reported in children born via CS. This may result from the lack of initial exposure to maternal vaginal and intestinal flora at birth, potentially leaving children vulnerable to various diseases due to alterations in immune system development [35].
Extensive research has focused on dietetic influence on infants’ gut microbiota, specifically on breast milk versus formulas [36]. Human milk (HM) oligosaccharides (HMOs) constitute the third most abundant component of breast milk, after lactose and lipids. By supplying these oligosaccharides, breastfeeding promotes the growth of specific Bifidobacterium and Lactobacillus species that utilize HMOs and dominate the ecosystem as long as the baby is predominantly breastfed [37]. HMOs are composed of glucose, galactose, N-acetylglucosamine, fucose, and sialic acid. From these five units, over 200 distinct HMO structures have been identified [37]. HMOs function as prebiotics by stimulating the growth of beneficial bacteria while preventing the binding of harmful bacteria to intestinal epithelial cells. Moreover, species in the gut that ferment sugars produce metabolites that may protect against infectious and immune-related diseases. Formula feeding, lacking HMOs and with higher protein content, promotes instead a diverse gut microbiota development with more opportunistic pathogens, such as E. coli, Clostridium, and Bacteroides, and a more proteolytic metabolism. However, as the child grows and changes the type of nutrition by introducing new foods, the gut microbiota becomes more diverse and stable, resembling that of an adult by around 3 to 5 years of age. The period between exclusive milk feeding and the transition to eating family foods is usually referred to as the complementary feeding (CF) period, typically spanning from 6 to 24 months of age [38]. The World Health Organization (WHO) recommends starting CF around 6 months of age. However, in certain countries, it is not uncommon to introduce other foods as early as 2 or 3 months of age [39]. Early initiation of CF, before 3 months of age, has been associated with various health risks that include increased susceptibility to GI and respiratory infections, obesity, and allergies. However, these risks might be more attributable to the shorter duration of breastfeeding rather than the introduction of complementary foods at an early age [40]. CF, by introducing dietary fibers and new protein sources, triggers a transformation in the gut microbiota—with increased Bacteroides, Lachnospiraceae, and Ruminococcaceae and a decreased presence of Bifidobacterium—and in its metabolism, transitioning from a milk-adapted state to a more mature and diverse adult-like community, characterized by higher levels of short-chain fatty acid-producing bacterial taxa [41].
In early life, particularly infancy and early childhood, the gut microbiota undergoes significant changes and is more susceptible to disruptions compared to adults (Figure 3) [39]. Moreover, a reduced overall diversity in gut microbiota during infancy has been linked to allergic diseases [36,42]. Based on recent advances, the use of probiotics, prebiotics, or fermented dairy products to manipulate gut microbiota has been proposed to treat or prevent various disorders, including intestinal dysbiosis [43], also in early life. In this review, therapeutic applications of pre- to postbiotic-related products, which have the potential to be used as an alternative to pharmacological agents in the treatment of pediatric intestinal dysbiosis associated with IBD, will be investigated.

2. Methods

A rigorous electronic search was conducted on well-known scientific databases, including PubMed, Scopus, and Web of Science. The search was limited to the last 10 years, from 2014 to June 2024. The search was performed using a combination of keywords including pediatric, *biotics, and dysbiosis (Figure 4). Boolean operators and filters have been applied to refine and optimize the search results, improving the accuracy and relevance of the results. In a second step, more detailed keywords, for example, SCFAs or butyric acid, were added to the search. The articles were assessed according to the title, abstract, and full text. Additional references were included through meticulous examination of the initially reviewed articles, ensuring broad inclusion of both recent and prior literature. The extracted data were then reviewed and synthesized using a narrative approach, providing a detailed and coherent summary of the current state of the research.

3. Prebiotics, Probiotics, Synbiotics, and Paraprobiotics: Their Effects on IBD Intestinal Dysbiosis Affecting Children

3.1. Prebiotics

The concept of prebiotics was first introduced by Glen Gibson and Marcel Roberfroid in 1995. An initial description defines them as non-digestible food ingredients with beneficial effects for the host by stimulating the activity and/or growth of one or a limited number of host bacteria in the colon [44]. Since then, the concept has been refined, but its core principles have remained the same. According to the most recent review of International Scientific Association for Probiotics and Prebiotics, the current definition for “dietary prebiotics” is “a selectively fermented ingredient that results in specific changes in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefit(s) upon host health” [45]. Furthermore, for an ingredient to be classified as prebiotic, it must exert its effect on a limited group of microorganisms and must comply with three aspects: be a substrate, have a physiologically beneficial effect, and have a microbiota-mediated mechanism. The effect exerted by prebiotics depends on their deployment by the host’s gut microbiota. This distinction separates prebiotics from other substances such as antibiotics, vitamins, bacteriophages, and minerals whose consumption alters gut microbiota composition.
Thus, prebiotics must have the following characteristics:
  • Indigestible by the host’s enzyme.
  • Fermented selectively by gastrointestinal microbiota.
  • Selectively promote the growth and/or activity of intestinal microbiota associated with health and well-being [45,46].
A large portion of prebiotics belongs to dietary fibers; nevertheless, not all have prebiotic properties. Some other main groups are inulin and fructo-oligosaccharide (FOS), galacto-oligosaccharides (GOS) including the family of raffinose oligosaccharides (RFO), trans-galacto-oligosaccharides (TOS), lactulose, resistant starch (RS), glucose-derived oligosaccharides such as polydextrose, and pectin oligosaccharides (POS). Prebiotics are naturally present in many plant-based foods, such as garlic, asparagus, beans, bananas, chicory roots, etc. [47,48], or synthetically produced to create food supplements [49]. Prebiotics reach the colon without being digested in the upper gastrointestinal tract [50] and exert their function by influencing gut microbiota through various mechanisms:
  • Fermentation: prebiotics are non-digestible but fermentable substrates that modulate bacterial taxa, enriching groups that utilize them [51].
  • Production of bioactive metabolites: the fermentation process yields bioactive metabolites such as SCFAs (butyrate, propionic acetic, and lactic acid) [52,53].
  • Modulation of bacterial composition: prebiotics stimulate beneficial bacterial species while potentially inhibiting harmful ones [54,55].
  • Cross-feeding: the metabolic action of prebiotics by some bacteria indirectly induces the growth of others [53,56,57].
  • Modulating gut environment: changes in pH levels of gut lumen as a consequence of fermentation products alter species composition [58].
Historically, Lactobacilli and Bifidobacteria, commonly known as probiotics, use prebiotics as substrates [44]. Data from high-throughput sequencing techniques reveal that prebiotics impact a broader range of microorganisms, but not all, in gut microbiota [56]. The gut microbiota acts as a homeostatic organ and participates in the fermentation of prebiotic substances, leading to the formation of bioactive metabolites and energy production with consequent beneficial effects on intestinal mucosa [59], protecting against pathogen colonization [60]. It is well-established in the literature that patients with intestinal dysbiosis associated with IBD exhibit increased colonization by harmful bacteria, which alters intestinal barrier permeability, giving rise to “leaky gut” syndrome and intestinal dysbiosis [61,62]. In this context, prebiotics contribute via the fermentation pathway, indirectly, to the enrichment of gut microflora with beneficent bacterial strains. These processes contribute to the formation of a microbiota that can overpower and/or resist intestinal dysbiosis and cause disease [63,64,65]. Moreover, the production of SCFAs as fermentation products of prebiotics also helps to maintain gut integrity [59,66].
Clinical studies on pediatric patients with IBD and the use of prebiotics are limited. HM is the first and an important source of prebiotics during pediatric nutrition. Studies demonstrate how the oligosaccharides contained in HM and HMOs, as described before, acting as prebiotic fermentable substrates, contribute to a healthy and balanced infant gut microbiota [67,68]. Newborn and infant supplementation with oligosaccharides to date is becoming usual due to the infant formula fortification and enrichment with these prebiotics, principally GOS, POS, and FOS [69,70,71]. However, formula fortification and enrichment with non-digestible carbohydrates is a research hot topic due to their beneficial effects on microbiota and gastrointestinal tract homeostasis and their functions in general [72,73]. Finally, SCFAs produced through fermentation of undigested oligosaccharides are associated with the maintenance of intestinal integrity and aid in blocking pathogens and toxins by binding to epithelial cells [74]. The correct development of the intestinal barrier during pediatric age is particularly important in premature infants, where there is a strong drive to develop intestinal dysbiosis and increased intestinal permeability, thereby increasing the risk of necrotizing enterocolitis [9].

3.2. Probiotics

The term probiotics is derived from a Greek word meaning “for life” and is used to define living non-pathogenic organisms and their derived beneficial effects on hosts. The current definition, established by the Food and Agriculture Organization (FAO) and the WHO in 2001 and slightly modified by the International Scientific Association for Probiotics and Prebiotics (ISAPP) in 2014 describes probiotics as “live microorganisms which, when administered in adequate amounts, confer a health benefit on the host” [75]. This emphasizes the viability of microorganisms as crucial for the classification of probiotics (Figure 5).
It is important to clarify the precise scope and optimal use of the term “probiotic” to ensure that scientists, industries, regulators, and consumers in the field of probiotics have a common understanding of what is currently known about probiotics, in line with the latest research. This clarification will facilitate the continued advancement of probiotic research and will guarantee that the benefits of probiotics are accurately conveyed to consumers and patients [75]. It is worth pointing out that the presence of live microorganisms in various foods and nutritional supplements is not uncommon. However, only those strains that have been subjected to rigorous scientific investigation and that have been found to exert a beneficial impact on human health can be considered probiotics. Microbes that are alive and present in traditional fermented foods and beverages typically do not meet the required evidence level for probiotics since their health effects have not been confirmed and the mixtures of microorganisms are largely uncharacterized. Fermented foods may be claimed as probiotics if they contain an adequately characterized live microorganism or strain at doses that may provide benefits documented by at least one positive human clinical trial conducted according to generally accepted scientific standards or, if applicable, local or national government recommendations and requirements. Finally, the microorganism must be safe [77]. The most prevalent genera of bacteria currently available as probiotics and exhibiting beneficial health effects include Bifidobacterium, Lactobacillus, Bacillus, Enterococcus, Pediococcus, and the yeast Saccharomyces [75,78,79].
Each genus contains numerous species, and each species comprises numerous strains. It is generally considered that the health effects of probiotics are strain-specific [80]. Probiotics are commercially available as single strains or in formulations in which more strains are present in different combinations [81]. It is therefore not feasible to define the mechanism by which probiotics exert their effects. Nevertheless, several common mechanisms have been identified. Probiotics can influence the composition of the gut microbiota, competing with pathogens for nutrients and binding sites on the intestinal wall, enhancing intestinal barrier function, and modulating the immune system [82]. Their capacity to produce antimicrobial substances and other metabolites such as SCFAs enables them to exert a direct or indirect effect on host health by lowering the pH [83]. In addition, these microorganisms have the capacity to affect the nervous system of the host through the gut–brain axis [84,85]. As evidenced by their ability to regulate the composition of the intestinal microbiota, probiotics have been implicated as a potential treatment option for a range of gastrointestinal conditions, including IBD, that lead to variations in the microbiota causing intestinal dysbiosis [46,86,87,88]. Probiotics have been significantly investigated regarding the management of inflammatory conditions. However, the evidence base in relation to the use of probiotics in the pediatric population with IBD is limited [89,90].
In accordance with the current evidence-based guidelines established by the European Crohn’s and Colitis Organisation (ECCO) and the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN), VSL#3 and Escherichia coli Nissle 1917 may be regarded as an efficacious treatment for maintenance in patients with ulcerative colitis [91]. However, this recommendation lacks sufficient evidence to be considered valid [92,93]. With respect to Crohn’s disease, the ECCO/ESPGHAN guidelines conclude that there is currently insufficient evidence to demonstrate that probiotics are beneficial for the induction or maintenance of remission. In summary, the available evidence indicates that selected probiotics may be useful in the treatment of ulcerative colitis but not in Crohn’s disease [94]. The clinical studies that support these conclusions and guidelines in the use of probiotics in pediatric IBD have recently been summarized [95,96]. To achieve the desired probiotic health effects, it is essential to administer probiotics in an adequate amount. The optimal dose of probiotics has not yet been clearly established and may vary depending on the specific probiotic strain, the condition being treated, and other factors. Until further data are available, it is advisable to utilize the established treatment regimen (probiotic dosage and formulation, duration of treatment), which has been demonstrated to be effective in well-designed and conducted randomized controlled trials (RCTs) for the specific indication under consideration [97].
Although there is evidence to support the use of specific probiotics in certain clinical contexts, further studies are frequently needed to confirm the effect of these probiotics, as well as to define the most suitable type, dose, and timing. As a general criterion, the administration of probiotics to children with no documented health benefits should not be recommended [98].

3.3. Synbiotics

Synbiotics are dietary supplements that combine probiotics and prebiotics in the form of synergism [99,100], increasing the beneficial effects of probiotics [101]. So, it should be reserved only for products in which the prebiotic compound selectively favors the probiotic organism [102]. According to the current understanding, there are two types of synbiotic approaches [103]:
  • Complementary, where the selection of the probiotic is based on beneficial impacts intended for the host, while the prebiotic is selected separately to enhance the levels of beneficial microbial components. The prebiotic can support the growth and activity of the probiotic but does so indirectly as part of its broader target spectrum.
  • Synergistic, in which the probiotic is again chosen based on specific beneficial effects on the host, but the prebiotic is chosen to stimulate specifically the growth and activity of the selected probiotic. In this case, the prebiotic is selected to have a higher affinity for the probiotic and is chosen to enhance its survival and growth in the host. It can also increase levels of microbiota beneficial in the host, but the main target is the ingested probiotic.
Studies have been conducted on the effects of synbiotics on the GI microbiota in infants using supplementation in infant formula [104,105]. At present, the available evidence is insufficient to support the regular inclusion of these “-biotics” in infant formulas for healthy or atopic infants who are unable to be breastfed and there is currently no strong evidence to suggest any benefits either. However, the use of synbiotics to treat intestinal dysbiosis in children remains theoretical. To the best of our knowledge, there is no documented use of synbiotics to treat intestinal dysbiosis in children.

3.4. Paraprobiotics

Paraprobiotics have been described using various terms, such as inactivated probiotics, ghost probiotics, and nonviable (dead) probiotics (Figure 6) [106]. Unlike traditional probiotics, which are live microorganisms, paraprobiotics include dead bacterial cells, cellular components, and cell lysate or microbial extracts with health benefits that are stable, safe for use, and confer advantages to the host [107]. As per the FAO/WHO, paraprobiotics are defined as inactivated (non-viable) microbial cells, which, when managed in sufficient amounts, present benefits to consumers [107]. Paraprobiotics can be used in various forms, including dietary supplements, functional foods, and pharmaceuticals. Research on paraprobiotics is still in its early stages, but studies have shown promising results in various health conditions, such as gastrointestinal disorders and inflammatory diseases [108]. To the best of our knowledge, like synbiotics, there is no documented use of paraprobiotics in the treatment of pediatric intestinal dysbiosis.

3.5. Next-Generation Probiotics

Next-Generation Probiotics (NGPs) refer to advanced formulations of probiotics that incorporate innovative approaches to enhance their effectiveness and specificity [110]. These formulations often include strains selected for their ability to survive stomach acid and bile salts, adhere to intestinal surfaces, and modulate gut microbiota composition. Additionally, they may include encapsulation technologies or prebiotics to improve delivery and viability. In this context, several commensal microbial species identified in the intestinal microbiota have been proposed as probiotics of NGPs. Among the most promising candidates are the strains of Akkermansia muciniphila species, Faecalibacterium prausnitzii (e.g., Fusobacterium prausnitzii), Anaerobutyricum hallii (e.g., Eubacterium hallii), as well as Bacteroides spp., Roseburia spp., Clostridium butyricum [111], B. pseudopodium, L. rhamnosus, L. acidophilus, and L. lactobacillus, which modulate the serotonergic system in IBD [112]. These commensal bacteria are associated with a healthy state of the intestinal microbiota when present in adequate quantities. The aim is to optimize health benefits by targeting specific health conditions or populations, such as those with gastrointestinal disorders or immune deficiencies. Research in this field continues to explore novel probiotic strains and delivery mechanisms to further improve efficacy and therapeutic outcomes. It can be stated that there is significant potential for NGPs to treat gastrointestinal disorders, such as IBD, that exert their therapeutic effects by outcompeting pathogenic bacteria, regulating gut motility, producing SCFAs, and improving gut permeability. However, further research is necessary to confirm the efficacy of NGPs in managing these conditions. As of today, the use of NGP in pediatrics is limited, and there is no application for the treatment of intestinal dysbiosis associated with IBD in the pediatric field.

4. Postbiotics, Metabiotics, Biogenics, or Simply Microbiota Metabolites: Beyond Probiotics and Prebiotics on Pediatric Intestinal Dysbiosis in IBD

Postbiotics, metabiotics, biogenics, inactivated microbial intact (nonviable) cells, inactivated probiotics, phantom probiotics, and microbiota metabolites are all the different names used for the “preparation of inanimate microorganisms and/or their components that confers a health benefit on the host” [108,113]. Postbiotics include SCFAs, tryptophan (Trp) metabolites, organic acids, bile acids, proteins, secreted enzymes, amino acids, neurotransmitters, vitamins, bacteriocins, terpenoids, biosurfactants, etc. [114,115]. Postbiotics have been defined as beneficial bioactive compounds originating from living organisms, commensal microbiota, in the gut environment during the nutrient fermentation process. With respect to living organisms (probiotics, synbiotics, etc.) [116] postbiotics possess some advantages in terms of stability, packaging, transportation, and storage. Knowing the specific structure of different postbiotics, they can be used in purified forms allowing for the elucidation of specific mechanisms of action and triggering only specific pathways. Moreover, the literature presents an application of postbiotic functional food production or the preparation of various commodities such as dairy foods, fish products, bread, vegetables, and meat due to their very low or potential non-toxicity [117,118]. A further advantage that makes them therapeutically attractive is the possibility to reach physiologically high concentrations together with their suitability for different administration ways following the principles of pharmacokinetics [119]. Their beneficial effects have been demonstrated not only on gut homeostasis and physiology, such as permeability, inflammation, oxidative damages, and immune response, but also on other tissues such as skin, oral mucosa, and the central nervous system (CNS) [120,121]. Finally, the safety profile of postbiotics over probiotics makes them suitable supplements also for newborns or infants, comprising the most vulnerable premature newborns or preterm newborns with compromised gut integrity and a high risk of probiotics sepsis [122,123,124]. Also in this regard, the postbiotic beneficial effects on infectious disease, inflammatory response [104,125], and gut morbidities such as colitis, enterocolitis, and necrotizing enterocolitis [118,126,127] have been demonstrated in newborns, infants, and children. From a biochemical point of view, the mechanistic aspects linked to the postbiotics biological effects are far from being elucidated. However, it is clear that numerous microbiota metabolites influence gut and brain functions such as metabolic and immune response or pain perception. A plethora of preclinical and clinical studies, indeed, support the concept of “ microbiota-gut-brain-axis ” and its physiological involvement in health and disease [128]. The gut–brain axis or more recently defined microbiota–gut–brain axis, is a complex communication system that links bidirectionally the gut and the nervous system comprising the central (CNS), autonomic (ANS), and enteric nervous systems (ENS) together with hormonal systems [129]. Increasing evidence supports the modulatory role of microbiota metabolites on different gut–brain axis pathways in IBD symptoms [130].

4.1. Short-Chain Fatty Acids (SCFAs)

SCFAs, the products of saccharolytic fermentation of complex carbohydrates by intestinal microorganisms, especially acetate (60%), propionate (25%), and butyrate (15%) (Figure 7), were confirmed to reduce intestinal pH value and inhibit pathogenic microorganisms playing distinct roles in maintaining gut health and overall homeostasis [131]. Some different mechanisms of action have been proposed for SCFA contribution to gut health comprising nutrient supply for epithelial cells (butyrate) [132], gut motility regulation [133], and gut barrier restoration [134]. Different preclinical studies performed on cell models allowed for the proposal of SCFAs as key regulators of the inflammation process, affecting the recruitment and transmigration of immune cells [135]. Moreover, some mechanistic hypotheses have been formulated on their modulation of endothelial and immune cell inflammation through the binding to different intra and extra-cellular receptors [136,137] and the consequent control of pro- (IL-10) or anti-inflammatory (IL1β, IL12, TNF-α, IL-8) cytokine production [138,139,140,141]. In certain cases, direct supplementation of SCFAs may be used to restore their levels in the gut, helping to manage intestinal dysbiosis and its associated conditions, emerging as potential IBD treatment [142]. SCFAs play a vital role in mitigating the adverse effects of intestinal dysbiosis by maintaining gut barrier integrity, reducing inflammation, modulating the immune system, regulating metabolic processes, and influencing the gut–brain axis. Their therapeutic potential is being explored through various dietary and supplementation strategies to restore gut health and overall homeostasis in IBD in adults [143,144] and pediatrics [145,146]. Partial or exclusive enteral nutrition, performed with formulas containing low saturated fat, heme, and taurine and high protein, were demonstrated to be effective in inducing remission in children with Crohn’s disease, but reduced SCFA levels were found [147,148,149]. Moreover, an ex vivo experiment performed on circulating mononuclear cells from IBD patients and healthy controls showed a reduced sensitivity of the circulating immune cells from IBD patients to the anti-inflammatory effects of butyrate [150]. All these data support the need to investigate mechanistic aspects that link diet, microbial changes, and microbiota metabolite changes, leading to a reduction of gut inflammation in IBD.
Various clinical studies report a reduction in SCFA stool concentration in children with different nutritional states [151,152,153], pathological conditions [154,155,156], or gastrointestinal disorders [157,158,159]. However, only two recent clinical studies have reported on the use of SCFAs as supplements or add-on therapies in IBD for adults [160] and children [161]. Conducted across three pediatric centers specializing in the diagnosis and treatment of pediatric intestinal dysbiosis associated with IBD, this pioneering study involved 72 pediatric patients with a median age of 13.5 years. Participants received either 150 mg of sodium butyrate or a placebo orally in capsule form every 12 h for a duration of 12 weeks. The study found that sodium butyrate supplementation, at a total daily dose of 300 mg, was not effective as an adjunctive treatment for children and adolescents with IBD [137].

4.2. Tryptophan Metabolites

Tryptophan (TRP), a precursor of important bioactive amine, is an essential amino acid that is necessarily obtained from the diet. In the gut, TRP is mainly transformed in kynurenine (KNY, 90%) and serotonin (3%), with the remaining 7% being metabolized by the microbiota in indole derivatives (Figure 7) [162,163,164]. A dysregulation of TRP metabolism has also been associated with different pathological conditions such as obesity, metabolic disorders [165,166], and IBD, especially in Crohn’s disease patients [167,168], leading to an increased KYN/TRP ratio. This dysregulation is also supported by cytokines and cortisol levels and gut microbiota variations in microorganism composition and number during gut and neuroinflammation [169,170]. Between KYN second metabolites we can mention kynurenic acid (KYNA), quinolinic acid (QA), and picolinic acid (PA) with different effects on immune and inflammatory response [171,172]. While QA shows neurotoxic and pro-inflammatory effects [173], PA and KYNA [174] instead demonstrated neuroprotectivity, both in the central and enteric nervous systems, and anti-inflammatory properties [175], modulating immune cell differentiation and function [176]. KYN/TRP ratio dysregulation, moreover, could be linked to IBD mood comorbidities such as anxiety and depression by the consequent dysregulation of the serotonin pathway (Figure 6) [162]. Recent literature, considering preclinical and clinical studies, supports the promotion of KYNA as a potential pharmacological tool for IBD [177,178]. Together, these considerations suggest an in-depth exploration of the possibility of promoting KYN derivatives as a target signaling pathway for IBD hyperalgesia, such as for inflammatory bowel syndrome [179]. Microbiota metabolism of the last 7% of TRP introduced with diet leads to the production of protective indole derivatives, such as indole3-propionic, -lactic, -acetic acids, indole-3 acetaldehyde, and indole acrylic acids (Figure 6) [164]. Indoles are signaling molecules produced by the microbiota to regulate bacteria physiology. Additionally, these TRP metabolites may also support gut immune and anti-inflammatory responses and epithelial barrier functions, mediated by the activation of the aryl hydrocarbon receptor (AhR) [180]. The downregulation of AhR and reduced production of its ligands has been observed in IBD and intestinal dysbiosis [181]. The therapeutic potential of TRP metabolites is still emerging in the literature, and for this reason, clinical trials are still lacking even more so for pediatric patients. However, recent studies investigated KYN, KYNA, and TRP metabolites in neuroinflammation [182], autism [183], Kawasaki disease [184], and early childhood adiposity [185].

5. Other Supplements

5.1. Polyunsaturated Fatty Acids

There is evidence that suggests a relationship between ω-3 polyunsaturated fatty acids (PUFAs) and the gut microbiota. ω-3 PUFAs may alter the diversity and abundance of the gut microbiome just as the gut microbiota may influence the metabolism and absorption of ω-3 PUFAs. In addition, imbalanced consumption of n-3/n-6 PUFAs can lead to gut dysbiosis. ω-3 PUFA deficiency disrupts the microbiota community in metabolic disorders. ω-3 PUFAs include docosahexaenoic acid (DHA, C22:6), eicosapentaenoic acid (EPA, C20:5), α-linolenic acid (ALA, C18:3), and docosapentaenoic acid (DPA, C22:5). ω-3 PUFAs, the main source of which is fish oil [186], have a significant influence on immune homeostasis and modulation of the gut microbiota [187]. Various studies [187,188,189,190] have found that supplemental PUFAs, such as DHA and EPA found in fish oil, improve diet-induced microbiome changes (along with improvements in lipid profiles and fatty liver disease), with supplementation having a bigger effect on restoring the intestinal microbiome homeostasis and increased short-chain fatty acid production, which is both beneficial to health and also indicative of positive changes to the microbiome. Kaliannan et al. [191] indicated the beneficial effects of ω-3 fatty acids on antibiotic-induced gut dysbiosis and obesity, suggesting ω-3 supplementation as a safe and effective method for preventing obesity in children exposed to antibiotics. In conclusion, PUFAs are pivotal in managing intestinal dysbiosis in children due to their anti-inflammatory properties, modulation of gut microbiota, enhancement of gut barrier function, and support of immune health.

5.2. Vitamin D

Vitamin D is a lipid-soluble vitamin that is absorbed from dietary sources or supplements in the proximal small intestine [192]. Recent studies have revealed the association between vitamin D deficiency and a multitude of diseases, including IBD [193,194], where vitamin D is intimately involved in the regulation of inflammation via a bidirectional relationship with the gut microbiota [195,196]. Studies also suggest that the amount of dietary vitamin D and its circulating levels may be involved in maintaining immune homeostasis in healthy individuals, partially via modulating the gut microbial composition [197]. Exclusively breastfed infants present a particular risk of vitamin D deficiency, due to its low concentration in breast milk [198], low maternal vitamin D levels, and daily intake, as well as the lack of exposure of newborns and suckling infants to sunlight [199]. Therefore, associations such as the American Academy of Pediatrics and ESPGHAN recommend vitamin D supplementation at doses of 400 IU/day in infants who are exclusively or partially breastfed [195,200]. Regarding vitamin D treatment in IBD pediatric patients, some clinical studies investigated the effectiveness and safety of vitamin D supplementation with the aim of restoring hypovitaminosis D associated with IBD [201,202]. To date, only a few studies investigated the beneficial effects of vitamin D supplementation in children with IBD, reporting a significant decrease in the IBD activity score and biochemical inflammatory markers, such as calprotectin and C reactive protein, and interleukins such as IL-2, IL17, and IL23 [203], but without any consideration regarding the effects on the intestinal microbiota.

5.3. Minerals: Zinc

The crucial role of zinc in intestinal function is well documented [204]. Zinc is considered a functional food for maintaining gastrointestinal mucosal function [205] and its deficiency significantly impacts the intestinal mucosa, leading to its degeneration and severe adverse effects. This deficiency causes thinning of the mucus layer and alterations in mucus composition that appear to be caused primarily by disrupted mucin synthesis at the post-translational level, as observed in both animal and human goblet cell studies [206,207]. Studies have shown that children with IBD often have lower levels of zinc compared to healthy peers. This deficiency can be due to malabsorption, increased intestinal loss, and inadequate dietary intake. Although zinc supplementation seems to offer potential benefits for managing these conditions in animal and cell models, investigations of its efficiency in pediatric IBD treatment are still lacking. Consequently, there are no specific guidelines for zinc supplementation in children with conditions like IBD [208]. Currently, there is limited well-documented information on the appropriate dosage for pediatric patients.

6. Other Therapies for Pediatric IBD

6.1. Fecal Microbiota Transplantation (FMT)

Fecal microbiota transplantation (FMT) has been suggested as a mechanism to restore intestinal bacterial diversity. FMT involves transferring stool from a healthy donor to a recipient with the aim of restoring the balance of gut microbiota and promoting therapeutic effects [209,210,211]. FMT can restore the balance and diversity of beneficial bacteria in the gut microbiota by aligning the recipient’s composition with that of the donor. The efficacy and safety of FMT in pediatric patients have been confirmed in several studies and has been applied for severe conditions such as IBD with presumed underlying intestinal dysbiosis, ulcerative colitis, and Crohn’s disease [212,213,214,215,216]. The difference between probiotic or prebiotic therapies and FMT lies in the breadth of bacterial strains transferred. While probiotics and prebiotics only supplement some bacterial strains, FMT offers a more comprehensive approach by transferring hundreds of strains at once. However, this broad spectrum of bacteria in FMT also poses potential risks. Donors might transfer opportunistic pathogenic bacteria or infections to recipients along with the beneficial strains. This risk underscores the importance of rigorous screening and testing protocols for both donors and recipients in FMT procedures to minimize adverse outcomes.

6.2. Vaginal Seeding

During vaginal delivery, newborns are naturally exposed to the mother’s vaginal and intestinal bacteria, which play a crucial role in the early development of the infant’s immune system and gut microbiome [217]. Infants born by CS miss this initial exposure and, in relation to vaginally delivered children, show altered microbiota development [218]. Emerging evidence suggests that cesarean delivery may be a risk factor for the later development of childhood problems such as metabolic and inflammatory diseases [219]. Vaginal microbial transfer, also known as vaginal seeding, is a process where infants born by CS are exposed to the mother’s vaginal microbiota, aiming to mimic the natural microbial exposure that occurs during vaginal birth. The process of vaginal seeding involves swabbing obtained from pregnant women just before delivery, stored and applied to newborns’ lips and then to their entire bodies [220] with a gauze or swab that has been previously incubated in the mother’s vaginal fluids. This is performed shortly after birth to introduce beneficial bacteria to the infant’s skin and mucous membranes, thereby promoting the development of a healthy microbiome. To modify the microbiome of a CS baby, researchers tested wiping these babies with swabs that had been in their mother’s vagina for about an hour before the cesarean birth [221]. They found that this technique could partially restore the microbiota of CS babies. Other researchers investigated the differences in the baby microbiome at birth and again at 6 weeks of age. They found that by 6 weeks, there were no significant differences in the microbiomes based on the mode of delivery [222]. As a result, they concluded that any clinical implications of microbiome differences at birth might be minimal or nonexistent. Concern exists about the potential transmission of pathogenic bacteria or viruses from mother to baby. To date, there is no evidence to support potential harm to the baby. The transmission of some pathogens that may be asymptomatic in the mother could cause serious adverse consequences for infants, including herpes simplex virus, Chlamydia trachomatis, Neisseria gonorrhea, and group B streptococcus. While the concept is promising, it is still under research, and healthcare professionals have yet to reach a consensus on its safety and efficacy. Vaginal seeding following a CS continues to be an experimental procedure and is not recommended as standard practice. There are currently no established guidelines for maternal screening, existing studies have been limited in scope, and there has been insufficient long-term follow-up to fully assess potential lasting effects.

7. Clinical Trials

We organized here, in Table 1, clinical trials with effects on intestinal microbiota cited all over the review, giving particular attention to the population enrolled, treatment, and principal outcomes.

8. Conclusions and Future Perspectives

In conclusion, the current body of evidence on the use of dietary supplements in pediatric intestinal dysbiosis remains limited. Although some studies suggest the benefits of probiotics and prebiotics in modulating the gut microbiota, further studies are needed to clarify the efficacy and safety of these interventions in the pediatric population. Additionally, the potential role of emerging supplements such as synbiotics (combinations of prebiotics and probiotics), paraprobiotics (non-viable microbial cells or cell fragments), and postbiotics (microbiota metabolites) warrants future investigations. These novel supplements may offer additional therapeutic options, but their effects and safety profiles need to be thoroughly explored in pediatric populations before they can be recommended for widespread use. Effective management of pediatric intestinal dysbiosis through dietary supplementation requires a comprehensive understanding of the underlying mechanisms of intestinal dysbiosis, the specific needs of the pediatric population, and the interactions of different types of supplements and the gut microbiota. Establishing well-defined clinical guidelines based on robust scientific evidence is crucial for ensuring the safe and effective use of these supplements in children. Overall, while there is potential in using dietary supplements to manage pediatric intestinal dysbiosis, the current evidence base is not yet sufficient to make definitive recommendations. Future research should focus on addressing these gaps to provide clearer guidance for healthcare providers and caregivers in the management of pediatric gut health.

Author Contributions

Writing—original draft preparation, P.C., R.O., O.X. and S.C.; writing—review and editing P.C., R.O., O.X. and S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Landman, C.; Quévrain, E. Le microbiote intestinal: Description, rôle et implication physiopathologique. Rev. Med. Interne 2016, 37, 418–423. [Google Scholar] [CrossRef] [PubMed]
  2. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  3. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef]
  4. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef]
  5. Selber-Hnatiw, S.; Rukundo, B.; Ahmadi, M.; Akoubi, H.; Al-Bizri, H.; Aliu, A.F.; Ambeaghen, T.U.; Avetisyan, L.; Bahar, I.; Baird, A.; et al. Human Gut Microbiota: Toward an Ecology of Disease. Front. Microbiol. 2017, 8, 1265. [Google Scholar] [CrossRef]
  6. Tamboli, C.P.; Neut, C.; Desreumaux, P.; Colombel, J.F. Dysbiosis in inflammatory bowel disease. Gut 2004, 53, 1–4. [Google Scholar] [CrossRef]
  7. Thapar, N.; Benninga, M.A.; Crowell, M.D.; Di Lorenzo, C.; Mack, I.; Nurko, S.; Saps, M.; Shulman, R.J.; Szajewska, H.; van Tilburg, M.A.L.; et al. Paediatric functional abdominal pain disorders. Nat. Rev. Dis. Primers 2020, 6, 89. [Google Scholar] [CrossRef] [PubMed]
  8. Sung, V.; D’Amico, F.; Cabana, M.D.; Chau, K.; Koren, G.; Savino, F.; Szajewska, H.; Deshpande, G.; Dupont, C.; Indrio, F.; et al. Lactobacillus reuteri to Treat Infant Colic: A Meta-analysis. Pediatrics 2018, 141, e20171811. [Google Scholar] [CrossRef] [PubMed]
  9. Baranowski, J.R.; Claud, E.C. Necrotizing Enterocolitis and the Preterm Infant Microbiome. Adv. Exp. Med. Biol. 2019, 1125, 25–36. [Google Scholar] [CrossRef]
  10. Zhang, S.; Qian, Y.; Li, Q.; Xu, X.; Li, X.; Wang, C.; Cai, H.; Zhu, J.; Yu, Y. Metabolic and Neural Mechanisms Underlying the Associations Between Gut Bacteroides and Cognition: A Large-Scale Functional Network Connectivity Study. Front. Neurosci. 2021, 15, 750704. [Google Scholar] [CrossRef]
  11. Saeed, N.K.; Al-Beltagi, M.; Bediwy, A.S.; El-Sawaf, Y.; Toema, O. Gut microbiota in various childhood disorders: Implication and indications. World J. Gastroenterol. 2022, 28, 1875–1901. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, B.-N.; Liu, X.-T.; Liang, Z.-H.; Wang, J.-H. Gut microbiota in obesity. World J. Gastroenterol. 2021, 27, 3837–3850. [Google Scholar] [CrossRef] [PubMed]
  13. Williams, B.L.; Hornig, M.; Parekh, T.; Lipkin, W.I. Application of novel PCR-based methods for detection, quantitation, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances. mBio 2012, 3, 10–1128. [Google Scholar] [CrossRef]
  14. Fattorusso, A.; Di Genova, L.; Dell’Isola, G.B.; Mencaroni, E.; Esposito, S. Autism Spectrum Disorders and the Gut Microbiota. Nutrients 2019, 11, 521. [Google Scholar] [CrossRef] [PubMed]
  15. Bajinka, O.; Tan, Y.; Abdelhalim, K.A.; Özdemir, G.; Qiu, X. Extrinsic factors influencing gut microbes, the immediate consequences and restoring eubiosis. AMB Expr. 2020, 10, 130. [Google Scholar] [CrossRef]
  16. Chiba, M.; Nakane, K.; Komatsu, M. Westernized Diet is the Most Ubiquitous Environmental Factor in Inflammatory Bowel Disease. Perm. J. 2019, 23, 18–107. [Google Scholar] [CrossRef] [PubMed]
  17. Racine, A.; Carbonnel, F.; Chan, S.S.M.; Hart, A.R.; Bueno-de-Mesquita, H.B.; Oldenburg, B.; van Schaik, F.D.M.; Tjønneland, A.; Olsen, A.; Dahm, C.C.; et al. Dietary Patterns and Risk of Inflammatory Bowel Disease in Europe: Results from the EPIC Study. Inflamm. Bowel Dis. 2016, 22, 345–354. [Google Scholar] [CrossRef]
  18. Sjögren, Y.M.; Jenmalm, M.C.; Böttcher, M.F.; Björkstén, B.; Sverremark-Ekström, E. Altered early infant gut microbiota in children developing allergy up to 5 years of age. Clin. Exp. Allergy 2009, 39, 518–526. [Google Scholar] [CrossRef]
  19. Hanski, I.; von Hertzen, L.; Fyhrquist, N.; Koskinen, K.; Torppa, K.; Laatikainen, T.; Karisola, P.; Auvinen, P.; Paulin, L.; Mäkelä, M.J.; et al. Environmental biodiversity, human microbiota, and allergy are interrelated. Proc. Natl. Acad. Sci. USA 2012, 109, 8334–8339. [Google Scholar] [CrossRef]
  20. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef]
  21. Sherriff, A.; Golding, J. Hygiene levels in a contemporary population cohort are associated with wheezing and atopic eczema in preschool infants. Arch. Dis. Child. 2002, 87, 26–29. [Google Scholar] [CrossRef] [PubMed]
  22. Pittayanon, R.; Lau, J.T.; Leontiadis, G.I.; Tse, F.; Yuan, Y.; Surette, M.; Moayyedi, P. Differences in Gut Microbiota in Patients With vs Without Inflammatory Bowel Diseases: A Systematic Review. Gastroenterology 2020, 158, 930–946.e1. [Google Scholar] [CrossRef] [PubMed]
  23. Kuenzig, M.E.; Fung, S.G.; Marderfeld, L.; Mak, J.W.Y.; Kaplan, G.G.; Ng, S.C.; Wilson, D.C.; Cameron, F.; Henderson, P.; Kotze, P.G.; et al. Twenty-first Century Trends in the Global Epidemiology of Pediatric-Onset Inflammatory Bowel Disease: Systematic Review. Gastroenterology 2022, 162, 1147–1159.e4. [Google Scholar] [CrossRef] [PubMed]
  24. Brockway, M. The role of antibiotic exposure and the effects of breastmilk and human milk feeding on the developing infant gut microbiome. Front. Public Health 2024, 12, 1408246. [Google Scholar] [CrossRef] [PubMed]
  25. Davis, E.C.; Castagna, V.P.; Sela, D.A.; Hillard, M.A.; Lindberg, S.; Mantis, N.J.; Seppo, A.E.; Järvinen, K.M. Gut microbiome and breast-feeding: Implications for early immune development. J. Allergy Clin. Immunol. 2022, 150, 523–534. [Google Scholar] [CrossRef] [PubMed]
  26. Biasucci, G.; Rubini, M.; Riboni, S.; Morelli, L.; Bessi, E.; Retetangos, C. Mode of delivery affects the bacterial community in the newborn gut. Early Hum. Dev. 2010, 86 (Suppl. S1), 13–15. [Google Scholar] [CrossRef] [PubMed]
  27. Yu, L.; Guo, Y.; Wu, J.-L. Influence of mode of delivery on infant gut microbiota composition: A pilot study. J. Obstet. Gynaecol. 2024, 44, 2368829. [Google Scholar] [CrossRef] [PubMed]
  28. Clarke, G.; Stilling, R.M.; Kennedy, P.J.; Stanton, C.; Cryan, J.F.; Dinan, T.G. Minireview: Gut microbiota: The neglected endocrine organ. Mol. Endocrinol. 2014, 28, 1221–1238. [Google Scholar] [CrossRef] [PubMed]
  29. Rutayisire, E.; Huang, K.; Liu, Y.; Tao, F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: A systematic review. BMC Gastroenterol. 2016, 16, 86. [Google Scholar] [CrossRef]
  30. Gomaa, E.Z. Human gut microbiota/microbiome in health and diseases: A review. Antonie Van Leeuwenhoek 2020, 113, 2019–2040. [Google Scholar] [CrossRef]
  31. Mueller, N.T.; Whyatt, R.; Hoepner, L.; Oberfield, S.; Dominguez-Bello, M.G.; Widen, E.M.; Hassoun, A.; Perera, F.; Rundle, A. Prenatal exposure to antibiotics, cesarean section and risk of childhood obesity. Int. J. Obes. 2015, 39, 665–670. [Google Scholar] [CrossRef] [PubMed]
  32. Kuhle, S.; Tong, O.S.; Woolcott, C.G. Association between caesarean section and childhood obesity: A systematic review and meta-analysis. Obes. Rev. 2015, 16, 295–303. [Google Scholar] [CrossRef] [PubMed]
  33. Adlercreutz, E.H.; Wingren, C.J.; Vincente, R.P.; Merlo, J.; Agardh, D. Perinatal risk factors increase the risk of being affected by both type 1 diabetes and coeliac disease. Acta Paediatr. 2015, 104, 178–184. [Google Scholar] [CrossRef] [PubMed]
  34. Black, M.; Bhattacharya, S.; Philip, S.; Norman, J.E.; McLernon, D.J. Planned Cesarean Delivery at Term and Adverse Outcomes in Childhood Health. JAMA 2015, 314, 2271–2279. [Google Scholar] [CrossRef] [PubMed]
  35. Neu, J.; Rushing, J. Cesarean versus vaginal delivery: Long-term infant outcomes and the hygiene hypothesis. Clin. Perinatol. 2011, 38, 321–331. [Google Scholar] [CrossRef]
  36. Laursen, M.F.; Bahl, M.I.; Michaelsen, K.F.; Licht, T.R. First Foods and Gut Microbes. Front. Microbiol. 2017, 8, 356. [Google Scholar] [CrossRef] [PubMed]
  37. Davis, J.C.C.; Totten, S.M.; Huang, J.O.; Nagshbandi, S.; Kirmiz, N.; Garrido, D.A.; Lewis, Z.T.; Wu, L.D.; Smilowitz, J.T.; German, J.B.; et al. Identification of Oligosaccharides in Feces of Breast-fed Infants and Their Correlation with the Gut Microbial Community. Mol. Cell. Proteom. 2016, 15, 2987–3002. [Google Scholar] [CrossRef] [PubMed]
  38. Castenmiller, J.; de Henauw, S.; Hirsch-Ernst, K.-I.; Kearney, J.; Knutsen, H.K.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; Pelaez, C.; et al. Appropriate age range for introduction of complementary feeding into an infant’s diet. EFSA J. 2019, 17, e05780. [Google Scholar] [CrossRef] [PubMed]
  39. Schiess, S.; Grote, V.; Scaglioni, S.; Luque, V.; Martin, F.; Stolarczyk, A.; Vecchi, F.; Koletzko, B. Introduction of complementary feeding in 5 European countries. J. Pediatr. Gastroenterol. Nutr. 2010, 50, 92–98. [Google Scholar] [CrossRef] [PubMed]
  40. Przyrembel, H. Timing of introduction of complementary food: Short- and long-term health consequences. Ann. Nutr. Metab. 2012, 60 (Suppl. S2), 8–20. [Google Scholar] [CrossRef]
  41. Laursen, M.F. Gut Microbiota Development: Influence of Diet from Infancy to Toddlerhood. Ann. Nutr. Metab. 2021, 77, 21–34. [Google Scholar] [CrossRef]
  42. Tanaka, M.; Nakayama, J. Development of the gut microbiota in infancy and its impact on health in later life. Allergol. Int. 2017, 66, 515–522. [Google Scholar] [CrossRef] [PubMed]
  43. Goulet, O. Potential role of the intestinal microbiota in programming health and disease. Nutr. Rev. 2015, 73 (Suppl. S1), 32–40. [Google Scholar] [CrossRef]
  44. Gibson, G.R.; Roberfroid, M.B. Dietary Modulation of the Human Colonic Microbiota: Introducing the Concept of Prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
  45. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  46. Selvamani, S.; Mehta, V.; Ali El Enshasy, H.; Thevarajoo, S.; El Adawi, H.; Zeini, I.; Pham, K.; Varzakas, T.; Abomoelak, B. Efficacy of Probiotics-Based Interventions as Therapy for Inflammatory Bowel Disease: A Recent Update. Saudi J. Biol. Sci. 2022, 29, 3546–3567. [Google Scholar] [CrossRef]
  47. Bamigbade, G.B.; Subhash, A.J.; Kamal-Eldin, A.; Nyström, L.; Ayyash, M. An Updated Review on Prebiotics: Insights on Potentials of Food Seeds Waste as Source of Potential Prebiotics. Molecules 2022, 27, 5947. [Google Scholar] [CrossRef] [PubMed]
  48. Kaur, A.P.; Bhardwaj, S.; Dhanjal, D.S.; Nepovimova, E.; Cruz-Martins, N.; Kuča, K.; Chopra, C.; Singh, R.; Kumar, H.; Șen, F.; et al. Plant Prebiotics and Their Role in the Amelioration of Diseases. Biomolecules 2021, 11, 440. [Google Scholar] [CrossRef] [PubMed]
  49. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef]
  50. Guarino, M.; Altomare, A.; Emerenziani, S.; Di Rosa, C.; Ribolsi, M.; Balestrieri, P.; Iovino, P.; Rocchi, G.; Cicala, M. Mechanisms of Action of Prebiotics and Their Effects on Gastro-Intestinal Disorders in Adults. Nutrients 2020, 12, 1037. [Google Scholar] [CrossRef]
  51. Gill, S.K.; Rossi, M.; Bajka, B.; Whelan, K. Dietary fibre in gastrointestinal health and disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 101–116. [Google Scholar] [CrossRef]
  52. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-Gut Microbiota Metabolic Interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  53. Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef] [PubMed]
  54. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [PubMed]
  55. Healey, G.; Murphy, R.; Butts, C.; Brough, L.; Whelan, K.; Coad, J. Habitual dietary fibre intake influences gut microbiota response to an inulin-type fructan prebiotic: A randomised, double-blind, placebo-controlled, cross-over, human intervention study. Br. J. Nutr. 2018, 119, 176–189. [Google Scholar] [CrossRef] [PubMed]
  56. Flint, H.J.; Duncan, S.H.; Scott, K.P.; Louis, P. Links between diet, gut microbiota composition and gut metabolism. Proc. Nutr. Soc. 2015, 74, 13–22. [Google Scholar] [CrossRef]
  57. Culp, E.J.; Goodman, A.L. Cross-feeding in the gut microbiome: Ecology and mechanisms. Cell Host Microbe 2023, 31, 485–499. [Google Scholar] [CrossRef] [PubMed]
  58. Duncan, S.H.; Louis, P.; Thomson, J.M.; Flint, H.J. The role of pH in determining the species composition of the human colonic microbiota. Environ. Microbiol. 2009, 11, 2112–2122. [Google Scholar] [CrossRef] [PubMed]
  59. Gonçalves, P.; Araújo, J.R.; Di Santo, J.P. A Cross-Talk Between Microbiota-Derived Short-Chain Fatty Acids and the Host Mucosal Immune System Regulates Intestinal Homeostasis and Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2018, 24, 558–572. [Google Scholar] [CrossRef] [PubMed]
  60. Bäumler, A.J.; Sperandio, V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature 2016, 535, 85–93. [Google Scholar] [CrossRef]
  61. Jin, Y.-T.; Duan, Y.; Deng, X.-K.; Lin, J. Prevention of necrotizing enterocolitis in premature infants—An updated review. WJCP 2019, 8, 23–32. [Google Scholar] [CrossRef] [PubMed]
  62. Ahmed, I.; Roy, B.; Khan, S.; Septer, S.; Umar, S. Microbiome, Metabolome and Inflammatory Bowel Disease. Microorganisms 2016, 4, 20. [Google Scholar] [CrossRef]
  63. Caballero-Flores, G.; Pickard, J.M.; Núñez, G. Microbiota-mediated colonization resistance: Mechanisms and regulation. Nat. Rev. Microbiol. 2023, 21, 347–360. [Google Scholar] [CrossRef]
  64. Darb Emamie, A.; Rajabpour, M.; Ghanavati, R.; Asadolahi, P.; Farzi, S.; Sobouti, B.; Darbandi, A. The effects of probiotics, prebiotics and synbiotics on the reduction of IBD complications, a periodic review during 2009–2020. J. Appl. Microbiol. 2021, 130, 1823–1838. [Google Scholar] [CrossRef]
  65. Naseer, M.; Poola, S.; Ali, S.; Samiullah, S.; Tahan, V. Prebiotics and Probiotics in Inflammatory Bowel Disease: Where are we now and where are we going? CCP 2020, 15, 216–233. [Google Scholar] [CrossRef]
  66. Roberfroid, M.B. Prebiotics: Preferential substrates for specific germs? Am. J. Clin. Nutr. 2001, 73, 406s–409s. [Google Scholar] [CrossRef]
  67. Hu, M.; Li, M.; Li, C.; Miao, M.; Zhang, T. Effects of Human Milk Oligosaccharides in Infant Health Based on Gut Microbiota Alteration. J. Agric. Food Chem. 2023, 71, 994–1001. [Google Scholar] [CrossRef]
  68. Chong, H.-Y.; Tan, L.T.-H.; Law, J.W.-F.; Hong, K.-W.; Ratnasingam, V.; Ab Mutalib, N.-S.; Lee, L.-H.; Letchumanan, V. Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022, 14, 3554. [Google Scholar] [CrossRef] [PubMed]
  69. Ben, X.-M.; Li, J.; Feng, Z.-T.; Shi, S.-Y.; Lu, Y.-D.; Chen, R.; Zhou, X.-Y. Low level of galacto-oligosaccharide in infant formula stimulates growth of intestinal Bifidobacteria and Lactobacilli. World J. Gastroenterol. 2008, 14, 6564–6568. [Google Scholar] [CrossRef] [PubMed]
  70. Prieto, P.A. In Vitro and Clinical Experiences with a Human Milk Oligosaccharide, Lacto-N- neoTetraose, and Fructooligosaccharides. Foods Food Ingred. J. Jpn. 2005, 210, 1018. [Google Scholar]
  71. Puccio, G.; Alliet, P.; Cajozzo, C.; Janssens, E.; Corsello, G.; Sprenger, N.; Wernimont, S.; Egli, D.; Gosoniu, L.; Steenhout, P. Effects of Infant Formula With Human Milk Oligosaccharides on Growth and Morbidity: A Randomized Multicenter Trial. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 624–631. [Google Scholar] [CrossRef] [PubMed]
  72. Verkhnyatskaya, S.; Ferrari, M.; de Vos, P.; Walvoort, M.T.C. Shaping the Infant Microbiome With Non-digestible Carbohydrates. Front. Microbiol. 2019, 10, 343. [Google Scholar] [CrossRef] [PubMed]
  73. Hu, A.; Chen, X.; Bi, Q.; Xiang, Y.; Jin, R.; Ai, H.; Nie, Y. A parallel and cascade control system: Magnetofection of miR125b for synergistic tumor-association macrophage polarization regulation and tumor cell suppression in breast cancer treatment. Nanoscale 2020, 12, 22615–22627. [Google Scholar] [CrossRef]
  74. Sun, W.; Tao, L.; Qian, C.; Xue, P.; Tong, X.; Yang, L.; Lu, F.; Wan, H.; Tao, Y. Human milk oligosaccharides and the association with microbiota in colostrum: A pilot study. Arch. Microbiol. 2024, 206, 58. [Google Scholar] [CrossRef]
  75. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  76. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and synbiotics—A review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef]
  77. Binda, S.; Hill, C.; Johansen, E.; Obis, D.; Pot, B.; Sanders, M.E.; Tremblay, A.; Ouwehand, A.C. Criteria to Qualify Microorganisms as “Probiotic” in Foods and Dietary Supplements. Front. Microbiol. 2020, 11, 1662. [Google Scholar] [CrossRef]
  78. Roy, S.; Dhaneshwar, S. Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: Current perspectives. World J. Gastroenterol. 2023, 29, 2078–2100. [Google Scholar] [CrossRef] [PubMed]
  79. Zendeboodi, F.; Khorshidian, N.; Mortazavian, A.M.; Da Cruz, A.G. Probiotic: Conceptualization from a new approach. Curr. Opin. Food Sci. 2020, 32, 103–123. [Google Scholar] [CrossRef]
  80. Maftei, N.-M.; Raileanu, C.R.; Balta, A.A.; Ambrose, L.; Boev, M.; Marin, D.B.; Lisa, E.L. The Potential Impact of Probiotics on Human Health: An Update on Their Health-Promoting Properties. Microorganisms 2024, 12, 234. [Google Scholar] [CrossRef]
  81. Collado, M.C.; Meriluoto, J.; Salminen, S. Development of new probiotics by strain combinations: Is it possible to improve the adhesion to intestinal mucus? J. Dairy Sci. 2007, 90, 2710–2716. [Google Scholar] [CrossRef] [PubMed]
  82. Kumar, R.; Sood, U.; Gupta, V.; Singh, M.; Scaria, J.; Lal, R. Recent Advancements in the Development of Modern Probiotics for Restoring Human Gut Microbiome Dysbiosis. Indian J. Microbiol. 2020, 60, 12–25. [Google Scholar] [CrossRef] [PubMed]
  83. Markowiak-Kopeć, P.; Śliżewska, K. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef] [PubMed]
  84. Mörkl, S.; Butler, M.I.; Holl, A.; Cryan, J.F.; Dinan, T.G. Probiotics and the Microbiota-Gut-Brain Axis: Focus on Psychiatry. Curr. Nutr. Rep. 2020, 9, 171–182. [Google Scholar] [CrossRef] [PubMed]
  85. Ansari, F.; Neshat, M.; Pourjafar, H.; Jafari, S.M.; Samakkhah, S.A.; Mirzakhani, E. The role of probiotics and prebiotics in modulating of the gut-brain axis. Front. Nutr. 2023, 10, 1173660. [Google Scholar] [CrossRef]
  86. Haneishi, Y.; Furuya, Y.; Hasegawa, M.; Picarelli, A.; Rossi, M.; Miyamoto, J. Inflammatory Bowel Diseases and Gut Microbiota. Int. J. Mol. Sci. 2023, 24, 3817. [Google Scholar] [CrossRef] [PubMed]
  87. Aggeletopoulou, I.; Konstantakis, C.; Assimakopoulos, S.F.; Triantos, C. The role of the gut microbiota in the treatment of inflammatory bowel diseases. Microb. Pathog. 2019, 137, 103774. [Google Scholar] [CrossRef] [PubMed]
  88. Štofilová, J.; Kvaková, M.; Kamlárová, A.; Hijová, E.; Bertková, I.; Guľašová, Z. Probiotic-Based Intervention in the Treatment of Ulcerative Colitis: Conventional and New Approaches. Biomedicines 2022, 10, 2236. [Google Scholar] [CrossRef] [PubMed]
  89. Tang, M.; Wang, C.; Xia, Y.; Tang, J.; Wang, J.; Shen, L. Clostridioides difficile infection in inflammatory bowel disease: A clinical review. Expert Rev. Anti Infect. Ther. 2024, 22, 297–306. [Google Scholar] [CrossRef]
  90. Siddiqui, A.; Haider, R.; Aaqil, S.I.; Vohra, L.I.; Qamar, K.; Jawed, A.; Fatima, N.; Adnan, A.; Parikh, V.; Ochani, S.; et al. Probiotic formulations and gastro-intestinal diseases in the paediatric population: A narrative review. Ann. Med. Surg. 2024, 86, 2836–2847. [Google Scholar] [CrossRef]
  91. Turner, D.; Levine, A.; Escher, J.C.; Griffiths, A.M.; Russell, R.K.; Dignass, A.; Dias, J.A.; Bronsky, J.; Braegger, C.P.; Cucchiara, S.; et al. Management of pediatric ulcerative colitis: Joint ECCO and ESPGHAN evidence-based consensus guidelines. J. Pediatr. Gastroenterol. Nutr. 2012, 55, 340–361. [Google Scholar] [CrossRef] [PubMed]
  92. Miele, E.; Pascarella, F.; Giannetti, E.; Quaglietta, L.; Baldassano, R.N.; Staiano, A. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am. J. Gastroenterol. 2009, 104, 437–443. [Google Scholar] [CrossRef]
  93. Huynh, H.Q.; de Bruyn, J.; Guan, L.; Diaz, H.; Li, M.; Girgis, S.; Turner, J.; Fedorak, R.; Madsen, K. Probiotic preparation VSL#3 induces remission in children with mild to moderate acute ulcerative colitis: A pilot study. Inflamm. Bowel Dis. 2009, 15, 760–768. [Google Scholar] [CrossRef] [PubMed]
  94. Ruemmele, F.M.; Veres, G.; Kolho, K.L.; Griffiths, A.; Levine, A.; Escher, J.C.; Amil Dias, J.; Barabino, A.; Braegger, C.P.; Bronsky, J.; et al. Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn’s disease. J. Crohns. Colitis 2014, 8, 1179–1207. [Google Scholar] [CrossRef] [PubMed]
  95. Corsello, A.; Scatigno, L.; Fiore, G.; Baresi, S.; Eletti, F.; Zuccotti, G.; Strisciuglio, C.; Dilillo, D.; Verduci, E. Nutraceuticals and biotics in pediatric gastrointestinal disorders. Eur. J. Clin. Nutr. 2024, 78, 87–98. [Google Scholar] [CrossRef] [PubMed]
  96. Guandalini, S.; Sansotta, N. Probiotics in the Treatment of Inflammatory Bowel Disease. Adv. Exp. Med. Biol. 2019, 1125, 101–107. [Google Scholar] [CrossRef] [PubMed]
  97. Szajewska, H. What are the indications for using probiotics in children? Arch. Dis. Child. 2016, 101, 398–403. [Google Scholar] [CrossRef]
  98. Szajewska, H.; Berni Canani, R.; Domellöf, M.; Guarino, A.; Hojsak, I.; Indrio, F.; Lo Vecchio, A.; Mihatsch, W.A.; Mosca, A.; Orel, R.; et al. Probiotics for the Management of Pediatric Gastrointestinal Disorders: Position Paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J. Pediatr. Gastroenterol. Nutr. 2023, 76, 232–247. [Google Scholar] [CrossRef] [PubMed]
  99. Akutko, K.; Stawarski, A. Probiotics, Prebiotics and Synbiotics in Inflammatory Bowel Diseases. J. Clin. Med. 2021, 10, 2466. [Google Scholar] [CrossRef]
  100. Steed, H.; Macfarlane, G.T.; Blackett, K.L.; Bahrami, B.; Reynolds, N.; Walsh, S.V.; Cummings, J.H.; Macfarlane, S. Clinical trial: The microbiological and immunological effects of synbiotic consumption—A randomized double-blind placebo-controlled study in active Crohn’s disease. Aliment. Pharmacol. Ther. 2010, 32, 872–883. [Google Scholar] [CrossRef]
  101. Jiménez-Villeda, B.E.; Falfán-Cortés, R.N.; Rangel-Vargas, E.; Santos-López, E.M.; Gómez-Aldapa, C.A.; Torres-Vitela, M.R.; Villarruel-López, A.; Castro-Rosas, J. Synbiotic Encapsulation: A Trend towards Increasing Viability and Probiotic Effect. J. Food Process. Preserv. 2023, 2023, 1–20. [Google Scholar] [CrossRef]
  102. Cencic, A.; Chingwaru, W. The role of functional foods, nutraceuticals, and food supplements in intestinal health. Nutrients 2010, 2, 611–625. [Google Scholar] [CrossRef] [PubMed]
  103. Kolida, S.; Gibson, G.R. Synbiotics in health and disease. Annu. Rev. Food Sci. Technol. 2011, 2, 373–393. [Google Scholar] [CrossRef]
  104. Lemoine, A.; Tounian, P.; Adel-Patient, K.; Thomas, M. Pre-, pro-, syn-, and Postbiotics in Infant Formulas: What Are the Immune Benefits for Infants? Nutrients 2023, 15, 1231. [Google Scholar] [CrossRef] [PubMed]
  105. Ferro, L.E.; Crowley, L.N.; Bittinger, K.; Friedman, E.S.; Decker, J.E.; Russel, K.; Katz, S.; Kim, J.K.; Trabulsi, J.C. Effects of prebiotics, probiotics, and synbiotics on the infant gut microbiota and other health outcomes: A systematic review. Crit. Rev. Food Sci. Nutr. 2023, 63, 5620–5642. [Google Scholar] [CrossRef] [PubMed]
  106. Siciliano, R.A.; Reale, A.; Mazzeo, M.F.; Morandi, S.; Silvetti, T.; Brasca, M. Paraprobiotics: A New Perspective for Functional Foods and Nutraceuticals. Nutrients 2021, 13, 1225. [Google Scholar] [CrossRef] [PubMed]
  107. Aguilar-Toalá, J.E.; Garcia-Varela, R.; Garcia, H.S.; Mata-Haro, V.; González-Córdova, A.F.; Vallejo-Cordoba, B.; Hernández-Mendoza, A. Postbiotics: An evolving term within the functional foods field. Trends Food Sci. Technol. 2018, 75, 105–114. [Google Scholar] [CrossRef]
  108. Monteiro, S.S.; Schnorr, C.E.; Pasquali, M.A.d.B. Paraprobiotics and Postbiotics-Current State of Scientific Research and Future Trends toward the Development of Functional Foods. Foods 2023, 12, 2394. [Google Scholar] [CrossRef] [PubMed]
  109. Cuevas-González, P.F.; Liceaga, A.M.; Aguilar-Toalá, J.E. Postbiotics and paraprobiotics: From concepts to applications. Food Res. Int. 2020, 136, 109502. [Google Scholar] [CrossRef]
  110. Abouelela, M.E.; Helmy, Y.A. Next-Generation Probiotics as Novel Therapeutics for Improving Human Health: Current Trends and Future Perspectives. Microorganisms 2024, 12, 430. [Google Scholar] [CrossRef]
  111. Dahiya, D.K.; Renuka; Dangi, A.K.; Shandilya, U.K.; Puniya, A.K.; Shukla, P. New-Generation Probiotics. In Microbiome and Metabolome in Diagnosis, Therapy, and other Strategic Applications; Elsevier: Amsterdam, The Netherlands, 2019; pp. 417–424. ISBN 9780128152492. [Google Scholar]
  112. Hizay, A.; Dag, K.; Oz, N.; Comak-Gocer, E.M.; Ozbey-Unlu, O.; Ucak, M.; Keles-Celik, N. Lactobacillus acidophilus regulates abnormal serotonin availability in experimental ulcerative colitis. Anaerobe 2023, 80, 102710. [Google Scholar] [CrossRef]
  113. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
  114. Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, Y.; Qin, S.; Jia, J.; Huang, L.; Li, F.; Jin, F.; Ren, Z.; Wang, Y. Intestinal Microbiota-Associated Metabolites: Crucial Factors in the Effectiveness of Herbal Medicines and Diet Therapies. Front. Physiol. 2019, 10, 1343. [Google Scholar] [CrossRef] [PubMed]
  116. Jang, H.J.; Lee, N.-K.; Paik, H.-D. A Narrative Review on the Advance of Probiotics to Metabiotics. J. Microbiol. Biotechnol. 2024, 34, 487–494. [Google Scholar] [CrossRef] [PubMed]
  117. Sharma, N.; Kang, D.-K.; Paik, H.-D.; Park, Y.-S. Beyond probiotics: A narrative review on an era of revolution. Food Sci. Biotechnol. 2023, 32, 413–421. [Google Scholar] [CrossRef] [PubMed]
  118. Nataraj, B.H.; Ali, S.A.; Behare, P.V.; Yadav, H. Postbiotics-parabiotics: The new horizons in microbial biotherapy and functional foods. Microb. Cell Fact. 2020, 19, 168. [Google Scholar] [CrossRef] [PubMed]
  119. Wong, A.C.; Levy, M. New Approaches to Microbiome-Based Therapies. mSystems 2019, 4, e00122-19. [Google Scholar] [CrossRef] [PubMed]
  120. Scott, E.; de Paepe, K.; van de Wiele, T. Postbiotics and Their Health Modulatory Biomolecules. Biomolecules 2022, 12, 1640. [Google Scholar] [CrossRef]
  121. Shen, G.; Wu, J.; Ye, B.-C.; Qi, N. Gut Microbiota-Derived Metabolites in the Development of Diseases. Can. J. Infect. Dis. Med. Microbiol. 2021, 2021, 6658674. [Google Scholar] [CrossRef]
  122. Morniroli, D.; Vizzari, G.; Consales, A.; Mosca, F.; Giannì, M.L. Postbiotic Supplementation for Children and Newborn’s Health. Nutrients 2021, 13, 781. [Google Scholar] [CrossRef]
  123. Wegh, C.A.M.; Geerlings, S.Y.; Knol, J.; Roeselers, G.; Belzer, C. Postbiotics and Their Potential Applications in Early Life Nutrition and Beyond. Int. J. Mol. Sci. 2019, 20, 4673. [Google Scholar] [CrossRef]
  124. Deshpande, G.; Athalye-Jape, G.; Patole, S. Para-probiotics for Preterm Neonates-The Next Frontier. Nutrients 2018, 10, 871. [Google Scholar] [CrossRef]
  125. Malagón-Rojas, J.N.; Mantziari, A.; Salminen, S.; Szajewska, H. Postbiotics for Preventing and Treating Common Infectious Diseases in Children: A Systematic Review. Nutrients 2020, 12, 389. [Google Scholar] [CrossRef]
  126. Zagato, E.; Mileti, E.; Massimiliano, L.; Fasano, F.; Budelli, A.; Penna, G.; Rescigno, M. Lactobacillus paracasei CBA L74 metabolic products and fermented milk for infant formula have anti-inflammatory activity on dendritic cells in vitro and protective effects against colitis and an enteric pathogen in vivo. PLoS ONE 2014, 9, e87615. [Google Scholar] [CrossRef]
  127. Athalye-Jape, G.; Rao, S.; Simmer, K.; Patole, S. Bifidobacterium breve M-16V as a Probiotic for Preterm Infants: A Strain-Specific Systematic Review. JPEN J. Parenter. Enter. Nutr. 2018, 42, 677–688. [Google Scholar] [CrossRef]
  128. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  129. Bistoletti, M.; Bosi, A.; Banfi, D.; Giaroni, C.; Baj, A. The microbiota-gut-brain axis: Focus on the fundamental communication pathways. Prog. Mol. Biol. Transl. Sci. 2020, 176, 43–110. [Google Scholar] [CrossRef]
  130. Banfi, D.; Moro, E.; Bosi, A.; Bistoletti, M.; Cerantola, S.; Crema, F.; Maggi, F.; Giron, M.C.; Giaroni, C.; Baj, A. Impact of Microbial Metabolites on Microbiota-Gut-Brain Axis in Inflammatory Bowel Disease. Int. J. Mol. Sci. 2021, 22, 1623. [Google Scholar] [CrossRef]
  131. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  132. Annison, G.; Topping, D.L. Nutritional role of resistant starch: Chemical structure vs. physiological function. Annu. Rev. Nutr. 1994, 14, 297–320. [Google Scholar] [CrossRef]
  133. Cherbut, C.; Ferrier, L.; Rozé, C.; Anini, Y.; Blottière, H.; Lecannu, G.; Galmiche, J.P. Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am. J. Physiol. 1998, 275, G1415–G1422. [Google Scholar] [CrossRef]
  134. Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef]
  135. Li, M.; van Esch, B.C.A.M.; Wagenaar, G.T.M.; Garssen, J.; Folkerts, G.; Henricks, P.A.J. Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharmacol. 2018, 831, 52–59. [Google Scholar] [CrossRef]
  136. Kim, S.; Kim, J.-H.; Park, B.O.; Kwak, Y.S. Perspectives on the therapeutic potential of short-chain fatty acid receptors. BMB Rep. 2014, 47, 173–178. [Google Scholar] [CrossRef]
  137. Davie, J.R. Inhibition of histone deacetylase activity by butyrate. J. Nutr. 2003, 133, 2485S–2493S. [Google Scholar] [CrossRef]
  138. Vinolo, M.A.R.; Rodrigues, H.G.; Hatanaka, E.; Sato, F.T.; Sampaio, S.C.; Curi, R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 2011, 22, 849–855. [Google Scholar] [CrossRef]
  139. Li, M.; van Esch, B.C.A.M.; Henricks, P.A.J.; Folkerts, G.; Garssen, J. The Anti-inflammatory Effects of Short Chain Fatty Acids on Lipopolysaccharide- or Tumor Necrosis Factor α-Stimulated Endothelial Cells via Activation of GPR41/43 and Inhibition of HDACs. Front. Pharmacol. 2018, 9, 533. [Google Scholar] [CrossRef]
  140. Weng, M.; Walker, W.A.; Sanderson, I.R. Butyrate regulates the expression of pathogen-triggered IL-8 in intestinal epithelia. Pediatr. Res. 2007, 62, 542–546. [Google Scholar] [CrossRef] [PubMed]
  141. Willemsen, L.E.M.; Koetsier, M.A.; van Deventer, S.J.H.; van Tol, E.A.F. Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E1 and E2 production by intestinal myofibroblasts. Gut 2003, 52, 1442–1447. [Google Scholar] [CrossRef] [PubMed]
  142. Rudiansyah, M.; Abdalkareem Jasim, S.; Azizov, B.S.; Samusenkov, V.; Kamal Abdelbasset, W.; Yasin, G.; Mohammad, H.J.; Jawad, M.A.; Mahmudiono, T.; Hosseini-Fard, S.R.; et al. The emerging microbiome-based approaches to IBD therapy: From SCFAs to urolithin A. J. Dig. Dis. 2022, 23, 412–434. [Google Scholar] [CrossRef]
  143. Sugihara, K.; Kamada, N. Diet-Microbiota Interactions in Inflammatory Bowel Disease. Nutrients 2021, 13, 1533. [Google Scholar] [CrossRef] [PubMed]
  144. Olendzki, B.; Bucci, V.; Cawley, C.; Maserati, R.; McManus, M.; Olednzki, E.; Madziar, C.; Chiang, D.; Ward, D.V.; Pellish, R.; et al. Dietary manipulation of the gut microbiome in inflammatory bowel disease patients: Pilot study. Gut Microbes 2022, 14, 2046244. [Google Scholar] [CrossRef] [PubMed]
  145. Healey, G.R.; Celiberto, L.S.; Lee, S.M.; Jacobson, K. Fiber and Prebiotic Interventions in Pediatric Inflammatory Bowel Disease: What Role Does the Gut Microbiome Play? Nutrients 2020, 12, 3204. [Google Scholar] [CrossRef]
  146. Alsharairi, N.A. The Therapeutic Role of Short-Chain Fatty Acids Mediated Very Low-Calorie Ketogenic Diet-Gut Microbiota Relationships in Paediatric Inflammatory Bowel Diseases. Nutrients 2022, 14, 4113. [Google Scholar] [CrossRef]
  147. Levine, A.; Wine, E.; Assa, A.; Sigall Boneh, R.; Shaoul, R.; Kori, M.; Cohen, S.; Peleg, S.; Shamaly, H.; On, A.; et al. Crohn’s Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology 2019, 157, 440–450.e8. [Google Scholar] [CrossRef] [PubMed]
  148. Matuszczyk, M.; Meglicka, M.; Landowski, P.; Czkwianianc, E.; Sordyl, B.; Szymańska, E.; Kierkuś, J. Oral exclusive enteral nutrition for induction of clinical remission, mucosal healing, and improvement of nutritional status and growth velocity in children with active Crohn’s disease—A prospective multicentre trial. Prz. Gastroenterol. 2021, 16, 346–351. [Google Scholar] [CrossRef]
  149. Niseteo, T.; Sila, S.; Trivić, I.; Mišak, Z.; Kolaček, S.; Hojsak, I. Modified Crohn’s disease exclusion diet is equally effective as exclusive enteral nutrition: Real-world data. Nutr. Clin. Pract. 2022, 37, 435–441. [Google Scholar] [CrossRef]
  150. Recharla, N.; Geesala, R.; Shi, X.-Z. Gut Microbial Metabolite Butyrate and Its Therapeutic Role in Inflammatory Bowel Disease: A Literature Review. Nutrients 2023, 15, 2275. [Google Scholar] [CrossRef]
  151. Surono, I.S.; Jalal, F.; Bahri, S.; Romulo, A.; Kusumo, P.D.; Manalu, E.; Yusnita; Venema, K. Differences in immune status and fecal SCFA between Indonesian stunted children and children with normal nutritional status. PLoS ONE 2021, 16, e0254300. [Google Scholar] [CrossRef]
  152. Śliżewska, K.; Włodarczyk, M.; Sobczak, M.; Barczyńska, R.; Kapuśniak, J.; Socha, P.; Wierzbicka-Rucińska, A.; Kotowska, A. Comparison of the Activity of Fecal Enzymes and Concentration of SCFA in Healthy and Overweight Children. Nutrients 2023, 15, 987. [Google Scholar] [CrossRef] [PubMed]
  153. Gyarmati, P.; Song, Y.; Dotimas, J.; Yoshiba, G.; Christison, A. Cross-sectional comparisons of gut microbiome and short-chain fatty acid levels among children with varied weight classifications. Pediatr. Obes. 2021, 16, e12750. [Google Scholar] [CrossRef] [PubMed]
  154. Holmes, Z.C.; Silverman, J.D.; Dressman, H.K.; Wei, Z.; Dallow, E.P.; Armstrong, S.C.; Seed, P.C.; Rawls, J.F.; David, L.A. Short-Chain Fatty Acid Production by Gut Microbiota from Children with Obesity Differs According to Prebiotic Choice and Bacterial Community Composition. mBio 2020, 11, e00914-20. [Google Scholar] [CrossRef] [PubMed]
  155. Cheng, H.Y.; Chan, J.C.Y.; Yap, G.C.; Huang, C.-H.; Kioh, D.Y.Q.; Tham, E.H.; Loo, E.X.L.; Shek, L.P.C.; Karnani, N.; Goh, A.; et al. Evaluation of Stool Short Chain Fatty Acids Profiles in the First Year of Life With Childhood Atopy-Related Outcomes. Front. Allergy 2022, 3, 873168. [Google Scholar] [CrossRef] [PubMed]
  156. Samuelsson, U.; Ludvigsson, J. The concentrations of short-chain fatty acids and other microflora-associated characteristics in faeces from children with newly diagnosed Type 1 diabetes and control children and their family members. Diabet. Med. 2004, 21, 64–67. [Google Scholar] [CrossRef] [PubMed]
  157. Ivanko, O.H.; Bilykh, V.M. Fecal concentrations of lactic acid and short-chain fatty acids in young children hospitalized in an infectious-diagnostic hospital with diarrhea. ZMJ 2022, 24, 332–337. [Google Scholar] [CrossRef]
  158. Demehri, F.R.; Frykman, P.K.; Cheng, Z.; Ruan, C.; Wester, T.; Nordenskjöld, A.; Kawaguchi, A.; Hui, T.T.; Granström, A.L.; Funari, V.; et al. Altered fecal short chain fatty acid composition in children with a history of Hirschsprung-associated enterocolitis. J. Pediatr. Surg. 2016, 51, 81–86. [Google Scholar] [CrossRef] [PubMed]
  159. Rotondo-Trivette, S.; Wang, B.; Luan, Y.; Fiehn, O.; Sun, F.; Michail, S. Reduced fecal short-chain fatty acids in hispanic children with ulcerative colitis. Physiol. Rep. 2021, 9, e14918. [Google Scholar] [CrossRef] [PubMed]
  160. Vernero, M.; de Blasio, F.; Ribaldone, D.G.; Bugianesi, E.; Pellicano, R.; Saracco, G.M.; Astegiano, M.; Caviglia, G.P. The Usefulness of Microencapsulated Sodium Butyrate Add-On Therapy in Maintaining Remission in Patients with Ulcerative Colitis: A Prospective Observational Study. J. Clin. Med. 2020, 9, 3941. [Google Scholar] [CrossRef]
  161. Pietrzak, A.; Banasiuk, M.; Szczepanik, M.; Borys-Iwanicka, A.; Pytrus, T.; Walkowiak, J.; Banaszkiewicz, A. Sodium Butyrate Effectiveness in Children and Adolescents with Newly Diagnosed Inflammatory Bowel Diseases-Randomized Placebo-Controlled Multicenter Trial. Nutrients 2022, 14, 3283. [Google Scholar] [CrossRef]
  162. Bosi, A.; Banfi, D.; Bistoletti, M.; Giaroni, C.; Baj, A. Tryptophan Metabolites Along the Microbiota-Gut-Brain Axis: An Interkingdom Communication System Influencing the Gut in Health and Disease. Int. J. Tryptophan Res. 2020, 13, 1178646920928984. [Google Scholar] [CrossRef] [PubMed]
  163. Dehhaghi, M.; Kazemi Shariat Panahi, H.; Guillemin, G.J. Microorganisms, Tryptophan Metabolism, and Kynurenine Pathway: A Complex Interconnected Loop Influencing Human Health Status. Int. J. Tryptophan Res. 2019, 12, 1178646919852996. [Google Scholar] [CrossRef] [PubMed]
  164. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef] [PubMed]
  165. Sas, K.; Szabó, E.; Vécsei, L. Mitochondria, Oxidative Stress and the Kynurenine System, with a Focus on Ageing and Neuroprotection. Molecules 2018, 23, 191. [Google Scholar] [CrossRef] [PubMed]
  166. Abenavoli, L.; Scarpellini, E.; Colica, C.; Boccuto, L.; Salehi, B.; Sharifi-Rad, J.; Aiello, V.; Romano, B.; de Lorenzo, A.; Izzo, A.A.; et al. Gut Microbiota and Obesity: A Role for Probiotics. Nutrients 2019, 11, 2690. [Google Scholar] [CrossRef] [PubMed]
  167. Nikolaus, S.; Schulte, B.; Al-Massad, N.; Thieme, F.; Schulte, D.M.; Bethge, J.; Rehman, A.; Tran, F.; Aden, K.; Häsler, R.; et al. Increased Tryptophan Metabolism Is Associated with Activity of Inflammatory Bowel Diseases. Gastroenterology 2017, 153, 1504–1516.e2. [Google Scholar] [CrossRef] [PubMed]
  168. Piqué, N.; Berlanga, M.; Miñana-Galbis, D. Health Benefits of Heat-Killed (Tyndallized) Probiotics: An Overview. Int. J. Mol. Sci. 2019, 20, 2534. [Google Scholar] [CrossRef] [PubMed]
  169. Gao, J.; Xu, K.; Liu, H.; Liu, G.; Bai, M.; Peng, C.; Li, T.; Yin, Y. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front. Cell. Infect. Microbiol. 2018, 8, 13. [Google Scholar] [CrossRef]
  170. Brooks, A.K.; Lawson, M.A.; Smith, R.A.; Janda, T.M.; Kelley, K.W.; McCusker, R.H. Interactions between inflammatory mediators and corticosteroids regulate transcription of genes within the Kynurenine Pathway in the mouse hippocampus. J. Neuroinflammation 2016, 13, 98. [Google Scholar] [CrossRef]
  171. Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef]
  172. Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 2017, 112, 399–412. [Google Scholar] [CrossRef]
  173. Lugo-Huitrón, R.; Ugalde Muñiz, P.; Pineda, B.; Pedraza-Chaverrí, J.; Ríos, C.; La Pérez-de Cruz, V. Quinolinic acid: An endogenous neurotoxin with multiple targets. Oxid. Med. Cell. Longev. 2013, 2013, 104024. [Google Scholar] [CrossRef]
  174. Grant, R.S.; Coggan, S.E.; Smythe, G.A. The physiological action of picolinic Acid in the human brain. Int. J. Tryptophan Res. 2009, 2, 71–79. [Google Scholar] [CrossRef] [PubMed]
  175. Dudzińska, E.; Szymona, K.; Kloc, R.; Gil-Kulik, P.; Kocki, T.; Świstowska, M.; Bogucki, J.; Kocki, J.; Urbanska, E.M. Increased expression of kynurenine aminotransferases mRNA in lymphocytes of patients with inflammatory bowel disease. Therap. Adv. Gastroenterol. 2019, 12, 1756284819881304. [Google Scholar] [CrossRef] [PubMed]
  176. Keszthelyi, D.; Troost, F.J.; Masclee, A.A.M. Understanding the role of tryptophan and serotonin metabolism in gastrointestinal function. Neurogastroenterol. Motil. 2009, 21, 1239–1249. [Google Scholar] [CrossRef]
  177. Desmons, A.; Humbert, L.; Eguether, T.; Krasniqi, P.; Rainteau, D.; Mahdi, T.; Kapel, N.; Lamazière, A. High performance liquid chromatography-tandem mass spectrometry quantification of tryptophan metabolites in human serum and stool—Application to clinical cohorts in Inflammatory Bowel Diseases. J. Chromatogr. A 2022, 1685, 463602. [Google Scholar] [CrossRef]
  178. Ye, Y.; Zhang, X.; Su, D.; Ren, Y.; Cheng, F.; Yao, Y.; Shi, G.; Ji, Y.; Chen, S.; Shi, P.; et al. Therapeutic efficacy of human adipose mesenchymal stem cells in Crohn’s colon fibrosis is improved by IFN-γ and kynurenic acid priming through indoleamine 2,3-dioxygenase-1 signaling. Stem Cell Res. Ther. 2022, 13, 465. [Google Scholar] [CrossRef]
  179. Clarke, G.; McKernan, D.P.; Gaszner, G.; Quigley, E.M.; Cryan, J.F.; Dinan, T.G. A Distinct Profile of Tryptophan Metabolism along the Kynurenine Pathway Downstream of Toll-Like Receptor Activation in Irritable Bowel Syndrome. Front. Pharmacol. 2012, 3, 90. [Google Scholar] [CrossRef] [PubMed]
  180. Stockinger, B.; Di Meglio, P.; Gialitakis, M.; Duarte, J.H. The aryl hydrocarbon receptor: Multitasking in the immune system. Annu. Rev. Immunol. 2014, 32, 403–432. [Google Scholar] [CrossRef]
  181. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  182. Yan, J.; Kothur, K.; Mohammad, S.; Chung, J.; Patel, S.; Jones, H.F.; Keating, B.A.; Han, V.X.; Webster, R.; Ardern-Holmes, S.; et al. CSF neopterin, quinolinic acid and kynurenine/tryptophan ratio are biomarkers of active neuroinflammation. eBioMedicine 2023, 91, 104589. [Google Scholar] [CrossRef] [PubMed]
  183. Kałużna-Czaplińska, J.; Jóźwik-Pruska, J.; Chirumbolo, S.; Bjørklund, G. Tryptophan status in autism spectrum disorder and the influence of supplementation on its level. Metab. Brain Dis. 2017, 32, 1585–1593. [Google Scholar] [CrossRef] [PubMed]
  184. Fan, X.; Li, K.; Guo, X.; Liao, S.; Zhang, Q.; Xu, Y.; Cui, H.; Zheng, L.; Xu, M. Metabolic profiling reveals altered tryptophan metabolism in patients with kawasaki disease. Front. Mol. Biosci. 2023, 10, 1180537. [Google Scholar] [CrossRef] [PubMed]
  185. Tan, K.M.-L.; Tint, M.-T.; Kothandaraman, N.; Michael, N.; Sadananthan, S.A.; Velan, S.S.; Fortier, M.V.; Yap, F.; Tan, K.H.; Gluckman, P.D.; et al. The Kynurenine Pathway Metabolites in Cord Blood Positively Correlate With Early Childhood Adiposity. J. Clin. Endocrinol. Metab. 2022, 107, e2464–e2473. [Google Scholar] [CrossRef]
  186. Lev-Tzion, R.; Griffiths, A.M.; Leder, O.; Turner, D. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst. Rev. 2014, 2014, CD006320. [Google Scholar] [CrossRef] [PubMed]
  187. Jayapala, H.P.S.; Lim, S.Y. N-3 Polyunsaturated Fatty Acids and Gut Microbiota. Comb. Chem. High Throughput Screen. 2023, 26, 892–905. [Google Scholar] [CrossRef] [PubMed]
  188. Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef] [PubMed]
  189. Zhang, Y.; Zhang, B.; Dong, L.; Chang, P. Potential of Omega-3 Polyunsaturated Fatty Acids in Managing Chemotherapy- or Radiotherapy-Related Intestinal Microbial Dysbiosis. Adv. Nutr. 2019, 10, 133–147. [Google Scholar] [CrossRef]
  190. Zhang, Z.; Zhang, H.; Chen, T.; Shi, L.; Wang, D.; Tang, D. Regulatory role of short-chain fatty acids in inflammatory bowel disease. Cell Commun. Signal. 2022, 20, 64. [Google Scholar] [CrossRef]
  191. Kaliannan, K.; Wang, B.; Li, X.-Y.; Bhan, A.K.; Kang, J.X. Omega-3 fatty acids prevent early-life antibiotic exposure-induced gut microbiota dysbiosis and later-life obesity. Int. J. Obes. 2016, 40, 1039–1042. [Google Scholar] [CrossRef]
  192. Hollander, D.; Truscott, T.C. Mechanism and site of small intestinal uptake of vitamin D3 in pharmacological concentrations. Am. J. Clin. Nutr. 1976, 29, 970–975. [Google Scholar] [CrossRef] [PubMed]
  193. Garg, M.; Hendy, P.; Ding, J.N.; Shaw, S.; Hold, G.; Hart, A. The Effect of Vitamin D on Intestinal Inflammation and Faecal Microbiota in Patients with Ulcerative Colitis. J. Crohns. Colitis 2018, 12, 963–972. [Google Scholar] [CrossRef]
  194. Nielsen, O.H.; Rejnmark, L.; Moss, A.C. Role of Vitamin D in the Natural History of Inflammatory Bowel Disease. J. Crohns. Colitis 2018, 12, 742–752. [Google Scholar] [CrossRef] [PubMed]
  195. Barengolts, E. Vitamin D and prebiotics may benefit the intestinal microbacteria and improve glucose homeostasis in prediabetes and type 2 diabetes. Endocr. Pract. 2013, 19, 497–510. [Google Scholar] [CrossRef] [PubMed]
  196. Cantorna, M.T.; Lin, Y.-D.; Arora, J.; Bora, S.; Tian, Y.; Nichols, R.G.; Patterson, A.D. Vitamin D Regulates the Microbiota to Control the Numbers of RORγt/FoxP3+ Regulatory T Cells in the Colon. Front. Immunol. 2019, 10, 1772. [Google Scholar] [CrossRef]
  197. Luthold, R.V.; Fernandes, G.R.; Franco-de-Moraes, A.C.; Folchetti, L.G.D.; Ferreira, S.R.G. Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals. Metabolism 2017, 69, 76–86. [Google Scholar] [CrossRef]
  198. við Streym, S.; Højskov, C.S.; Møller, U.K.; Heickendorff, L.; Vestergaard, P.; Mosekilde, L.; Rejnmark, L. Vitamin D content in human breast milk: A 9-mo follow-up study. Am. J. Clin. Nutr. 2016, 103, 107–114. [Google Scholar] [CrossRef]
  199. Tan, M.L.; Abrams, S.A.; Osborn, D.A. Vitamin D supplementation for term breastfed infants to prevent vitamin D deficiency and improve bone health. Cochrane Database Syst. Rev. 2020, 12, CD013046. [Google Scholar] [CrossRef]
  200. Wagner, C.L.; Greer, F.R. Prevention of rickets and vitamin D deficiency in infants, children, and adolescents. Pediatrics 2008, 122, 1142–1152. [Google Scholar] [CrossRef]
  201. Lee, R.; Maltz, R.M.; Crandall, W.V.; Plogsted, S.W.; Shaikhkhalil, A.K.; Bowden, S.A.; Mezoff, E.A. Single High-dose Vitamin D3 Supplementation in Pediatric Patients with Inflammatory Bowel Disease and Hypovitaminosis D. J. Pediatr. Gastroenterol. Nutr. 2020, 70, e77–e80. [Google Scholar] [CrossRef]
  202. Simek, R.Z.; Prince, J.; Syed, S.; Sauer, C.G.; Martineau, B.; Hofmekler, T.; Freeman, A.J.; Kumar, A.; McElhanon, B.O.; Schoen, B.T.; et al. Pilot Study Evaluating Efficacy of 2 Regimens for Hypovitaminosis D Repletion in Pediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2016, 62, 252–258. [Google Scholar] [CrossRef] [PubMed]
  203. El Amrousy, D.; El Ashry, H.; Hodeib, H.; Hassan, S. Vitamin D in Children with Inflammatory Bowel Disease: A Randomized Controlled Clinical Trial. J. Clin. Gastroenterol. 2021, 55, 815–820. [Google Scholar] [CrossRef]
  204. Semrad, C.E. Zinc and intestinal function. Curr. Gastroenterol. Rep. 1999, 1, 398–403. [Google Scholar] [CrossRef]
  205. Duggan, C.; Gannon, J.; Walker, W.A. Protective nutrients and functional foods for the gastrointestinal tract. Am. J. Clin. Nutr. 2002, 75, 789–808. [Google Scholar] [CrossRef]
  206. Quarterman, J.; Jackson, F.A.; Morrison, J.N. The effect of zinc deficiency on sheep intestinal mucin. Life Sci. 1976, 19, 979–986. [Google Scholar] [CrossRef] [PubMed]
  207. Maares, M.; Keil, C.; Straubing, S.; Robbe-Masselot, C.; Haase, H. Zinc Deficiency Disturbs Mucin Expression, O-Glycosylation and Secretion by Intestinal Goblet Cells. Int. J. Mol. Sci. 2020, 21, 6149. [Google Scholar] [CrossRef]
  208. Chao, H.-C. Zinc Deficiency and Therapeutic Value of Zinc Supplementation in Pediatric Gastrointestinal Diseases. Nutrients 2023, 15, 4093. [Google Scholar] [CrossRef] [PubMed]
  209. Chen, C.-C.; Chiu, C.-H. Current and future applications of fecal microbiota transplantation for children. Biomed. J. 2022, 45, 11–18. [Google Scholar] [CrossRef]
  210. Gu, X.; Chen, Z.-H.; Zhang, S.-C. Fecal microbiota transplantation in childhood: Past, present, and future. World J. Pediatr. 2023, 19, 813–822. [Google Scholar] [CrossRef]
  211. Elgarten, C.W.; Margolis, E.B.; Kelly, M.S. The Microbiome and Pediatric Transplantation. J. Pediatr. Infect. Dis. Soc. 2024, 13, S80–S89. [Google Scholar] [CrossRef]
  212. Karolewska-Bochenek, K.; Grzesiowski, P.; Banaszkiewicz, A.; Gawronska, A.; Kotowska, M.; Dziekiewicz, M.; Albrecht, P.; Radzikowski, A.; Lazowska-Przeorek, I. A Two-Week Fecal Microbiota Transplantation Course in Pediatric Patients with Inflammatory Bowel Disease. Adv. Exp. Med. Biol. 2018, 1047, 81–87. [Google Scholar] [CrossRef] [PubMed]
  213. Kunde, S.; Pham, A.; Bonczyk, S.; Crumb, T.; Duba, M.; Conrad, H.; Cloney, D.; Kugathasan, S. Safety, tolerability, and clinical response after fecal transplantation in children and young adults with ulcerative colitis. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 597–601. [Google Scholar] [CrossRef] [PubMed]
  214. Caldeira, L.d.F.; Borba, H.H.; Tonin, F.S.; Wiens, A.; Fernandez-Llimos, F.; Pontarolo, R. Fecal microbiota transplantation in inflammatory bowel disease patients: A systematic review and meta-analysis. PLoS ONE 2020, 15, e0238910. [Google Scholar] [CrossRef]
  215. Hourigan, S.K.; Chen, L.A.; Grigoryan, Z.; Laroche, G.; Weidner, M.; Sears, C.L.; Oliva-Hemker, M. Microbiome changes associated with sustained eradication of Clostridium difficile after single faecal microbiota transplantation in children with and without inflammatory bowel disease. Aliment. Pharmacol. Ther. 2015, 42, 741–752. [Google Scholar] [CrossRef] [PubMed]
  216. Hsu, M.; Tun, K.M.; Batra, K.; Haque, L.; Vongsavath, T.; Hong, A.S. Safety and Efficacy of Fecal Microbiota Transplantation in Treatment of Inflammatory Bowel Disease in the Pediatric Population: A Systematic Review and Meta-Analysis. Microorganisms 2023, 11, 1272. [Google Scholar] [CrossRef] [PubMed]
  217. DuPont, H.L.; Salge, M.M.H. The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health. Antibiotics 2023, 12, 1617. [Google Scholar] [CrossRef] [PubMed]
  218. Dunn, A.B.; Jordan, S.; Baker, B.J.; Carlson, N.S. The Maternal Infant Microbiome: Considerations for Labor and Birth. MCN Am. J. Matern. Child Nurs. 2017, 42, 318–325. [Google Scholar] [CrossRef] [PubMed]
  219. Liu, S.; Luo, X.; Zhou, L.; Xie, R.-H.; He, Y. Microbiota transplantation in restoring cesarean-related infant dysbiosis: A new frontier. Gut Microbes 2024, 16, 2351503. [Google Scholar] [CrossRef] [PubMed]
  220. Wilson, B.C.; Butler, É.M.; Grigg, C.P.; Derraik, J.G.B.; Chiavaroli, V.; Walker, N.; Thampi, S.; Creagh, C.; Reynolds, A.J.; Vatanen, T.; et al. Oral administration of maternal vaginal microbes at birth to restore gut microbiome development in infants born by caesarean section: A pilot randomised placebo-controlled trial. EBioMedicine 2021, 69, 103443. [Google Scholar] [CrossRef]
  221. Dominguez-Bello, M.G.; de Jesus-Laboy, K.M.; Shen, N.; Cox, L.M.; Amir, A.; Gonzalez, A.; Bokulich, N.A.; Song, S.J.; Hoashi, M.; Rivera-Vinas, J.I.; et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat. Med. 2016, 22, 250–253. [Google Scholar] [CrossRef]
  222. Chu, D.M.; Ma, J.; Prince, A.L.; Antony, K.M.; Seferovic, M.D.; Aagaard, K.M. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 2017, 23, 314–326. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Factors causing alteration to gut microbiota.
Figure 1. Factors causing alteration to gut microbiota.
Microorganisms 12 01582 g001
Figure 2. From in utero to post-natal life: factors that have been shown to affect the establishment of the gut microbiota.
Figure 2. From in utero to post-natal life: factors that have been shown to affect the establishment of the gut microbiota.
Microorganisms 12 01582 g002
Figure 3. Change in gut microbiota with aging: dominant bacterial species found in human intestines during different stages of growth, from infancy to adolescence.
Figure 3. Change in gut microbiota with aging: dominant bacterial species found in human intestines during different stages of growth, from infancy to adolescence.
Microorganisms 12 01582 g003
Figure 4. Principal keywords used for the literature search.
Figure 4. Principal keywords used for the literature search.
Microorganisms 12 01582 g004
Figure 5. Characteristics of an ideal probiotic strain [76]. GRAS = Generally Recognized as Safe.
Figure 5. Characteristics of an ideal probiotic strain [76]. GRAS = Generally Recognized as Safe.
Microorganisms 12 01582 g005
Figure 6. Conceptualization of paraprobiotics and postbiotics [109].
Figure 6. Conceptualization of paraprobiotics and postbiotics [109].
Microorganisms 12 01582 g006
Figure 7. Short-chain fatty acids and tryptophan metabolites.
Figure 7. Short-chain fatty acids and tryptophan metabolites.
Microorganisms 12 01582 g007
Table 1. Clinical trials.
Table 1. Clinical trials.
Reference PopulationTreatmentEffects
Ben, X. et al. [69]Prebiotics371 Term infants Infants 4 weeks after birth randomly assigned to IF or IF + GOS for 3 monthsHM and IF + GOS
intestinal Bifidobacteria, Lactobacilli Microorganisms 12 01582 i001
Fecal pH Microorganisms 12 01582 i002
Prieto, P.A. et al. [70]Prebiotics84 Term infants Infants within 11 days after birth, randomly assigned to IF or IF + FOS for 16 weeksHM and IF + FOS
Intestinal Lactobacilli Microorganisms 12 01582 i001
Puccio, G. et al. [71]Prebiotics175 Term infants Infants within 14 days after birth, randomly assigned to IF or IF + 20FL + LNnT for 6 monthsIF + 20FL + LNnT
safe and well tolerated,
morbidity (bronchitis) and medication Microorganisms 12 01582 i002
use (antipyretics and antibiotics)
Miele, E. et al. [92]Probiotics29 patients (mean age: 9.8 years; female/male: 13/16),Children newly diagnosed for UC, randomly assigned to VSL#3 weight-based dose,
(range: 450–1800 billion bacteria/day) (n = 14) or placebo (n = 15) *
Endoscopic, histological scores Microorganisms 12 01582 i002
No biochemical or clinical adverse events
Huynh, H.Q. et al. [93]Probiotics18 patients (mean age: 12.2 years; female/male: 7/11),All UC patients received 3 g sachet of VSL#3 twice daily by mouth for 8 Weeks10 patients remission (SCCAI < 3);
1 patient response (decrease in SCCAI 2, final score 5);
7 patients no changes.
Bacterial taxonomy changes
VSL#3 well tolerated
No adverse effects
Pietrzak, A. et al. [161]Postbiotics72 patients (mean age: 13.5 years; female/male: 14/28): 42 Crohn’s disease, 30 mild conditionsRandomly assigned to sodium butyrate 150 mg twice a day for 12 weeks (n = 29) or placebo (n = 23)Not effective as an adjunctive treatment
Kunde, S. et al. [213]FMT9 patients (7–21 years)
Mild to moderate UC
Freshly prepared fecal enemas daily for 5 days7 patients response within 1 week,
6 patients maintained response at 1 month.
No adverse effects, good tolerability
Wilson, B.C. et al. [220]Vaginal seeding47 newborns
22 Vaginal delivery (control)
12 Cesarean-seeded
13 Cesarean placebo
Newborns randomized to 3 mL solution of maternal vaginal microbes or sterile water; stool samples at 1 h, 1 month, and 3 months undergoing shotgun metagenomic sequencingNo differences in gut microbiome composition or functional potential were observed
HM (Human milk), IF (Infant Formula), FOS (fructooligosaccharide), GOS (galacto-oligosaccharides), 20FL (1.0 g/L 20fucosyllactose), LNnT (0.5 g/L lacto-N-neotetraose), * Concomitant steroid induction and mesalamine maintenance treatment for both groups, Microorganisms 12 01582 i001 = increase, Microorganisms 12 01582 i002 = decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ottria, R.; Xynomilakis, O.; Casati, S.; Ciuffreda, P. Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases. Microorganisms 2024, 12, 1582. https://doi.org/10.3390/microorganisms12081582

AMA Style

Ottria R, Xynomilakis O, Casati S, Ciuffreda P. Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases. Microorganisms. 2024; 12(8):1582. https://doi.org/10.3390/microorganisms12081582

Chicago/Turabian Style

Ottria, Roberta, Ornella Xynomilakis, Silvana Casati, and Pierangela Ciuffreda. 2024. "Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases" Microorganisms 12, no. 8: 1582. https://doi.org/10.3390/microorganisms12081582

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop