Next Article in Journal
Obstructive Sleep Apnea Is Associated with Worsened Hospital Outcomes in Children Hospitalized with Asthma
Next Article in Special Issue
The Role of Diet and Physical Activity in Obesity and Overweight in Children with Down Syndrome in Developed Countries
Previous Article in Journal
The Relationship between Maternal Smoking and Infant Birth Weight: Improving Accuracy through Urine Cotinine Analysis and Effective Medical Record Strategies
Previous Article in Special Issue
Iron and Vitamin A Status of Children Aged 0 to 36 Months in Thulamela Municipality, Vhembe District, South Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances

by
Lourdes Herrera-Quintana
1,2,†,
Héctor Vázquez-Lorente
1,2,†,
Daniel Hinojosa-Nogueira
3 and
Julio Plaza-Diaz
4,5,6,*
1
Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain
2
Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
3
Unidad de Gestión Clínica de Endocrinología y Nutrición, Laboratorio del Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario de Málaga (Virgen de la Victoria), 29590 Málaga, Spain
4
Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
5
Instituto de Investigación Biosanitaria IBS, GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
6
Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Children 2024, 11(8), 1030; https://doi.org/10.3390/children11081030
Submission received: 30 July 2024 / Revised: 16 August 2024 / Accepted: 20 August 2024 / Published: 22 August 2024
(This article belongs to the Special Issue Infant and Early Childhood Nutrition)

Abstract

:
Childhood is a critical period for immune system development, which is greatly influenced by the gut microbiome. Likewise, a number of factors affect the gut microbiome composition and diversity, including breastfeeding, formula feeding, and solid foods introduction. In this regard, several studies have previously demonstrated that breastfeeding promotes a favorable microbiome. In contrast, formula feeding and the early incorporation of certain solid foods may adversely affect microbiome development. Additionally, there is increasing evidence that disruptions in the early microbiome can lead to allergic conditions and food intolerances. Thus, developing strategies to promote optimal infant nutrition requires an understanding of the relationship between infant nutrition and long-term health. The present review aims to examine the relationship between infant feeding practices and the microbiome, as well as its implications on allergies and food intolerances in infants. Moreover, this study synthesizes existing evidence on how different eating habits influence the microbiome. It highlights their implications for the prevention of allergies and food intolerances. In conclusion, introducing allergenic solid foods before six months, alongside breastfeeding, may significantly reduce allergies and food intolerances risks, being also associated with variations in gut microbiome and related complications.

1. Introduction

1.1. Overview of Infant Feeding Practices and Recommendations

Healthy eating practices during the first years of a child’s life are essential for adequate development and growth [1]. Therefore, dietary patterns should be shaped during infancy to ensure a lifetime of health and well-being. However, the World Health Organization (WHO) estimates that undernutrition accounts for 2.7 million (45%) of all child deaths annually [2]. Primarily, the first two years are particularly crucial, with ideal nutrition during this period lowering morbidity and mortality, decreasing the risk of chronic diseases, and enhancing their overall development [3].
Globally, infant nutrition guidelines face significant barriers. The economic environment, such as poverty, can limit the availability of health care, food, and other resources essential to mothers and infants’ health [4]. Geographic isolation makes critical resources unavailable. A mother’s need to work outside of the home, the availability of family support networks, access to community healthcare workers, and her autonomy also affect infant nutrition [5]. Moreover, cultural factors also influence breastfeeding and other food habits [6].
In general terms, breastfeeding should be initiated within one hour of birth, being the exclusive feeding source during the first 6 months of life. After this period, the introduction of complementary foods (solids) is recommended along with continuing breastfeeding until 2 years of age or beyond, as proposed by WHO and United Nations International Children’s Emergency Fund (UNICEF) [2,7].

1.1.1. Breastfeeding

It is widely acknowledged that breastfeeding is the best food source for infants, guaranteeing a child’s well-being and health [8]. In addition to being clean, safe, and containing antibodies that help protect against many common childhood illnesses, it is also highly nutritious [9]. During the first 6 months of a baby’s life, breastmilk provides all the energy and nutrients they need. Furthermore, throughout the first 6 months to one year of life, it continues to meet 50% to 70% of their nutritional requirements [10]. However, contrary to official recommendations, the exclusive breastfeeding of infants under 6 months of age is estimated to be fewer than 50% [8]. Breastfeeding practice has been associated with several benefits for both mother and infant. For instance, as a result of breastfeeding, children are more likely to perform well on intellect tests, and less probable to be overweight or obese [11] and to develop diabetes later in life [2,8,9,10]. On the other hand, breastfeeding is linked to reduced risk of breast and ovarian cancer in women [2,8,9,10].

1.1.2. Formula Feeding

The infant formula may serve as an alternative to human milk as the unique basis of nutrition for infants (i.e., children under the age of 12 months) [12]. These formulas are designed in such a manner as to preserve a balance between macro- and micronutrients. There are three main types of infant formulas: usual infant formula (0–6 months), continuation formula (6–12 months), and toddler formula (13–36 months). Both large and small populations of mothers who are unable to breastfeed their infants are served by the infant formula [12]. Generally, the majority of guidelines recommend that infants who are formula-fed should use commercial infant formula until they reach 12 months of age, and that infant formula is not necessary beyond that age [13]. Regarding cow milk, it is recommended delaying its use until 9–12 months in order to reduce iron deficiency in children [14].

1.1.3. Introduction of Solid Foods

Complementary foods introduction to infants is a critical step in their development and long-term health status [15,16]. Additionally, there is evidence that the order in which complementary foods are introduced, their variety, and repeated exposure to these foods contribute to the development of food preferences later in life [17,18,19]. According to the European Food and Safety Agency (EFSA), no precise age for complemental feeding can be established, as it is strongly influenced by the infant’s characteristics and development [20]. According to WHO, infant formula is considered a complementary food, which is not directly comparable with the recommendations of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) and EFSA, both excluding infant formula as a complementary food [20,21].
Solid food introduction at the appropriate period may be a modifiable factor to improve a child’s growth and nutritional status [22,23]. Approximately after the first 6 months of life, infant formula or breast milk is no longer sufficient to meet the infant’s energy and nutrient requirements, and hence, solid foods should be initiated [24]. The introduction of solid food very early (at or before 4 months) appears to be related to a higher body mass index (BMI) at an older age [25,26,27,28,29]. According to the American Academy of Pediatrics (AAP), the Canadian Pediatric Society, and the WHO, solid food should be introduced at the age of 6 months, while according to ESPGHAN, it should be introduced between the ages of 4–6 months [30,31,32,33,34,35].

1.2. Importance of the Gut Microbiome and Its Early Development

Approximately 500–1000 species are thought to comprise the microbiota or microbiome, which includes a full collection of microbes that live in a particular biological niche, including bacteria, viruses, and fungi [36,37,38,39]. Microbes in the gut influence several aspects of human health, including immune and metabolic functions [40,41], or neurobehavioral characteristics [42]. As part of the microbiome of the gut, host nutrients, xenobiotics, and drugs are metabolized, the intestinal mucosal barrier is maintained, immune responses are modulated, and protection from pathogens is provided [43]. Moreover, a healthy gut microbiome plays an essential role in the fermentation of nondigestible substrates such as endogenous intestinal mucus and dietary fibers. In this process, short chain fatty acids (SCFAs) and gases are produced by selective microbes [44], with acetate, propionate, and butyrate being the most commonly produced SCFAs [45].
The composition of the gut microbiome is influenced by several factors. At the early stages, some of these factors include the method of delivery (i.e., vaginal or caesarean), the diet during infancy (e.g., formula or breast milk feeds), or the use of antibiotics [46,47]. This development and composition of the human gut microbiome has garnered significant attention owing to the crucial function of microbes in maintaining host health [48]. Early development of the intestinal microbiome determines adult microbiome composition and, therefore, health [49].
Infant microbiome composition can be traced to any of the maternal source communities on average across all infants [50]. There are multiple niches for infants in all maternal source communities. The microbiome of infants is shaped by shared and niche-specific host/environment factors [50]. In this regard, and according to longitudinal studies, the microbial structure of infants changes significantly with the cessation of feeding (breast or formula) and, therefore, with the introduction of solid foods [51]. As a result of the weaning process, the gut microbiome composition becomes stable as the taxonomic groups shift and the gut microbiome diversity increases from an infant to an adult. This process is modulated by diet [48,52]. Upon incorporating solid foods into the diet, the microbiome evolves from a simple environment, populated by Bifidobacterium (microorganisms that digest human milk oligosaccharides (HMOs)), to one that is populated by Bacteroides, which can metabolize starches present in a complex dietary pattern [53]. Thus, during the first years of life, the interaction between host and microbiome plays an important role, because considerable changes in structure and abundance happen at this important stage of the infant’s development [49].
Based on the above, the present review aims to examine the relationship between infant feeding practices and the microbiome, as well as its implications on allergies and food intolerances in infants.

2. Infant Feeding and the Microbiome Development

2.1. The Human Milk Microbiome: Composition and Impact on the Organism

The composition of the human milk microbiome may be determined by many factors, including lactation stage, maternal BMI, age, and diet [54], as well as by parity, geographical location, socioeconomic status, antibiotic or probiotic use during pregnancy, and type of delivery [54,55]. Among the nine genera that comprise the “core” bacteriome of the human milk microbiome are Streptococcus, Staphylococcus, Pseudomonas, Serratia, Corynebacterium, Ralstonia, Sphingomonas, Bradyrhizobium, and Propionibacterium. Approximately half of the microbial community in milk is represented by them, although the abundance of these bacteria varies from one sample to another [56,57].
In addition, it must be highlighted that breast milk also contains prebiotics, immunological substances, and other microbiome-shaped compounds to provide the infant with its own microbiome, possibly affecting colonization patterns elsewhere in the body [58]. As a result of the human milk microbiome supplying nutrients and dictating the production of metabolites, children’s gut and respiratory microbiomes are shaped [58,59,60].
Compared to the skin and respiratory microbiome, the fecal microbiome shows a continuing buildup of microbial diversity, while the skin and respiratory microbiome typically exhibit a unimodal pattern with a high initial diversity of microbes that rapidly declines over the first week, followed by a gradual increase in diversity [50]. Previous research suggests that the respiratory and skin microbiome, which are known as low-biomass places, may be seeded by other (low density) sources in addition to maternal seeding (e.g., other caregivers, hospitals, etc.) [61,62,63,64,65].

2.2. Benefits of Breastfeeding on Gut Health

Evidence highlights the importance of the “window of opportunity” during early childhood as being crucial to the gut microbiome long-term [66]. Therefore, the composition and metabolism of the neonate’s gut microbiome may play a critical role in the development of allergies [67]. In early life, breastfeeding affects the gut microbiome by exposing the neonate to the milk microbiome, both directly and indirectly, by incorporating factors in the maternal milk such as HMOs, secretory immunoglobulin (Ig)-A, and antimicrobials [68].
According to previous studies, the diversity of fecal bacteria increases with age, indicating that the microbial community becomes more complex with time [62,69]. Exclusively breastfed infants have shown to have a lower microbiome diversity than formula-fed babies, who present a higher diversity in their gut microbiome, being more similar to that of older children [70,71,72]. Therefore, breastfeeding results in a low level of bacterial diversity due to the predominance of infant-type Bifidobacterium, which is beneficial for children’s health. Infants who are exposed to Bifidobacterium in their early years may experience fewer infections due to the healthy maturation of their immune systems [73]. Likewise, Bacteroides has been seen to be another beneficial bacterium during the neonatal period since it plays a significant and specific role in the mucosal immune system development [74]. The presence of Bacteroides is also related to an earlier maturation rate and increased gut diversity [75].
Additionally, it is possible that other components of human milk may mediate the infant gut microbiome when interacting with the human milk microbiome. Secretory IgA, for example, may neutralize or inhibit the colonization of the human milk microbiome within the infant gut; this association remains unclear at present [76]. A study has shown that approximately 40% of human milk bacteria are IgA-coated [77], which may stimulate their colonization in the infant’s digestive system [76,78]. Human milk bacteria and other microorganisms, such as bacteriophages, remain largely unknown regarding their functional capabilities [79,80].
Although fewer studies have been conducted, human milk has been demonstrated to have a substantial impact on both the taxonomic composition (species abundance) and the functional capacity (functional related genes) of the resident microbiome, as well as their output (metabolites) [81]. Based on studies utilizing shotgun metagenomic sequencing, which sequences all genetic material within a microbial community, breastfeeding has been related to an enrichment in functional genes involved in fatty acid biosynthesis, vitamin B synthesis, and oxidative phosphorylation [52,62]. Infants’ microbiomes who do not receive human milk, on the other hand, exhibit a greater abundance of functional genes involved in methanogenesis and bile acid synthesis [62], as well as amino acid and nucleotide metabolism [52], all of which are typical of the more developed microbiome in adults.

2.3. Formula Feeding: Implications in Microbiome and Related Possibilities of Formulation

Interestingly, many advances have been made in the composition of formulas to mimic breast milk. In addition to identifying more constituents of human milk and describing their physiological functions, novel formulas continue to be developed, and established formulas are constantly being modified [82]. As a result, infant formula is increasingly supplemented with functional ingredients that are also found in breast milk to enhance its health-promoting capabilities [12,83,84], such as docosahexaenoic acid, lactoferrin, vitamins, minerals, and others. Despite this, infant formula cannot contain immunological components, enzymes, or hormones found in breast milk [82].
On the other hand, infant formulas can be designed to modulate the gut microbiome by incorporating probiotics, prebiotics, and symbiotics. The predominant probiotic strains incorporated into infant formulas are derived from the fecal microbiome of infants [85]. In this line, probiotics like Bifidobacterium and Lactobacillus [86], or oligosaccharides (prebiotics), may selectively stimulate the growth or metabolic activity of potentially beneficial indigenous bacteria such as bifidobacteria [87]. Furthermore, Streptococcus and Propionibacterium represent additional promising strains for use as probiotics [85]. It is critical to note that ESPGHAN posits that the administration of formulas fortified with probiotics and/or prebiotics currently used in infant formulas does not give rise to concerns regarding growth or possible adverse effects [88].

2.4. Timing and Types of First Foods and Their Impact on Microbiome Diversity

In the first 1 to 3 years of life, the gut microbiome of an infant undergoes significant changes from a relatively simple niche with low richness and diversity to one that resembles that of an adult [62]. Currently, we do not know how the introduction of complementary food (diversity of diet and food choices), after a diet consisting exclusively of milk, specifically affects the structure, diversity, and taxonomy of gut microbes [89,90]. Introducing solid foods may lead to the development of a microbiome that resembles that of an adult. This is a result of the changing ratios of protein, fat, fiber, and carbohydrate in the diet [91]. The observation of these gut bacteria in the adult population suggests that the introduction of solid foods facilitates the maturation of the gut microbiome of the child, leading to a configuration similar to that of adults [92].
During exclusive breast milk or formula feeding, the infant’s intestinal microbiome is predominantly composed of bifidobacteria and enterococci. Following the introduction of solid foods into the infant’s diet, there is a notable decline in the levels of these microorganism, while the relative abundance of fiber fermenters, including Lachnospiraceae, Bacteroidaceae, and Ruminococcaceae, increases [93]. In a recent study, infants’ gut microbiome was found to differ greatly between individuals at the time of introduction of solid food [94]. A strong cohort effect was also observed, which was expected, since the composition of gut microbiome is identified to vary geographically [95]. Based on the literature, inter-individual variability decreased with increasing age after the introductory period [62]. Within the first few weeks following birth to one year of age (14 months in the Netherlands), alpha diversity has been shown to increase over time [62,96]. Alpha diversity refers to the diversity of the microbiome in a single sample. In terms of alpha diversity, there are a number of indices that reflect different aspects of the heterogeneity of a community [97]. Thus, multifactorial components affect the gut microbiome at an early age, which may have a crucial role determining the microbiome’s composition and health in later life.

3. Microbiome and Allergies

3.1. Allergies: Underlying Mechanisms and Links with the Microbiome

Allergy pathogenesis involves a variety of factors, including genetics, epigenetics, environmental factors, microecology, and the immune system [98]. It is believed that a wide variety of cytokines, chemokines, allergens, and microbial products contribute to the complex cellular processes mediated by the adaptative and innate immune systems [99]. Particularly, a significant increase in Ig-E is associated with allergies, which are defined by a T-helper 2 (Th2) hypersensitivity response [100,101]. This immune response is characterized by the expression of cytokines, such as interleukin (IL)-13, IL-5, IL-4, and IL-9, leading to an inflammatory response [100,101]. In this line, regulatory T cells (Tregs) have been extensively studied and treated as potential therapeutic tools [102,103], since they display phenotypic and functional differences based on tissue location, differentiation status, disease state, and activation [104,105,106,107], enabling them to have diverse roles in allergy pathogenesis, among other processes [108,109].
For its part, the intestinal epithelium has a critical function in maintaining bodily homeostasis by serving as a protective barrier between the internal organs and the luminal microbiome [110,111]. Mucosal inflammation is characterized by the disruption of the intestinal epithelial barrier. In this regard, an increased exposure to luminal microbes triggers a perpetuating inflammatory response [112]. As a result of inflammation of the intestinal mucosa, multiple mechanisms may contribute to junctional disassembly and disruption of the epithelial barrier [113,114,115,116]. Among these mechanisms are a decreased expression of various tight junction (TJ) and adherens junction proteins, an impaired vesicular trafficking of junctional components, or an altered assembly and contractility of junctional actomyosin cytoskeletons [113,114,116,117].
A number of variables can affect intestinal permeability, including the modification of the gut microbiome, epithelial damage, dietary factors, and mucus layer dysfunction [118]. A dysbiosis, which may be caused by environmental factors, including drugs or diet, impairs the function of the epithelial barrier and produces proinflammatory cytokines. As a result of this process, TJ integrity is damaged and gut permeability is increased, resulting in leaky gut syndrome [119]. Thus, gut microbiome dysbiosis, in addition to affecting the intestinal epithelial barrier integrity, can result in antigen entry into the bloodstream and abnormal immune system stimulation [120]. Hence, intestinal and systemic diseases have been associated with impaired intestinal barrier function [121]. This may explain, in part, the relationship between respiratory allergies and gut microbiome dysbiosis [122].
Furthermore, other links between the gut microbiome and immune system have been reported in the literature. For instance, the gut-associated lymphoid tissue is a complex network of immune cells located at the deepest section of gut barrier [123]. Dendritic cells (DCs), intraepithelial DCs, intraepithelial lymphocytes, macrophages, TCD4+ lymphocytes, Tregs, plasma cells, and B lymphocytes are contained within the lamina propria [123]. Another example is the differentiation of T-helper 1 (Th1) cells, which is induced by contact with bacteria in the early stages of development. Hence, and taking all the above into consideration, it has been postulated that allergies could be prevented by regulating the intestinal microbiome [100,101,124].

3.2. Childhood Allergies and Microbiome

Over the past few decades, childhood allergies have become an increasingly prevalent issue [125], with various environmental factors, apart from genetic predisposition, potentially contributing to their development, such as cesarean birth, dietary habits, and antibiotic use. These factors also have an influence on the gut microbiome [126,127]. Moreover, it is known that dysbiosis, characterized by an imbalance between the function and composition of the microbiome, results in a breakdown of gut homeostasis and the development of diseases [128].
According to previous reports, allergic children have a higher number of Bacillota taxa and a lower number of Bacteroidota taxa [129]. Furthermore, an increased abundance of Faecalibacterium prausnitzii, Ruminococcus gnavus, Blautia wexlerae, and Anaerostipes hadrus has been observed in children’s gut microbiome with respiratory allergies and food allergies, while Bifidobacterium longum, Bacteroides dorei, Bacteroides vulgatus, Ruminococcus bromii, and several other fiber-degrading species have been found to be significantly lower than in healthy controls [130,131,132,133,134,135]. Figure 1 represents the main changes in the gut microbiome during infant feeding and in relation to allergies.
On the other hand, evidence shows that microbe metabolites, particularly SCFAs formed after microbial fermentation of undigested fiber, are critical for maintaining epithelial integrity and stimulating immune tolerance [136,137]. Several studies have demonstrated that allergic subjects have low fecal levels of SCFAs [138,139,140], while children whose microbiomes lack genes related to fiber fermentation have a greater risk of developing allergic sensitization [130]. Moreover, a reduction in the ability to catabolize polysaccharides and the production of pro-inflammatory molecules may be contributing factors to allergic inflammation [127].
The relationship between microbiome and allergies may be mediated by a number of mechanisms; one of those mechanisms is immune system modulation, wherein the gut microbiome can produce anti-inflammatory cytokines, such as IL-10, and induce Tregs to modulate the immune system [141,142]. The Tregs are responsible for maintaining immune tolerance to harmless antigens, such as food proteins. As a result of a disturbed gut microbiome, individuals may experience decreased levels of Tregs and anti-inflammatory cytokines, resulting in a decrease in immune tolerance and the development of food allergies [143]. A further potential mechanism previously mentioned involves the gut microbiome, which contributes to maintaining the gut barrier function and prevents harmful substances from entering the bloodstream [144].

3.3. Microbiome Signature in Food Allergies

It is widely accepted that alterations in gut bacteria levels or diversity are responsible for the recent rise in food allergy rates [145,146]. During digestion and absorption, allergens from food are absorbed in the gut, primarily in the small intestine, which comprises a symbiotic microbiome [147]. As humans and the microbiome evolve together, symbiotic microbes will inescapably have a significant influence on human health. Because of the intestinal cavity’s high microbial diversity, immune cells react positively to allergens in this environment [148]. The human intestinal microbiome, particularly those in the ileum and colon, has been demonstrated to play a significant role in mucosal immunity in the intestinal tract through the promotion of local homeostasis and the regulation of the immune response to food allergens [149].
Recently, several landmark studies have revealed a microbiome signature in food allergies, particularly among children, which suggests that dysbiosis during childhood may predict the persistence of the disorder. Food allergy risk has been demonstrated to be influenced by the early colonization of the gut microbiome in babies [146]. It has been demonstrated that the intestinal microbiome composition of infants aged 3 to 6 months affects their milk allergy resolution at the age of 8 years. As compared with the children whose milk allergy persists, the children whose milk allergy resolves later have an intestinal bacterial composition that is enriched in Clostridia and Bacillota after birth [150].
As a result of consuming milk formula for 6 months, infants with cow’s milk allergies have a higher total bacteria count, particularly anaerobic bacteria [151]. In addition, there were higher levels of lactobacilli and lower levels of Enterobacteriaceae and Bifidobacterium in babies with cow’s milk allergies [151]. According to Bunyavanich et al. [150], Clostridia and Bacillota were particularly high in gut microbiome of infants whose cow’s milk allergy had solved by the age of eight [150]. Moreover, Fazlollahi et al. [152] found that the gut microbiomes of children suffering from egg allergy were more abundant with genera associated with Lachnospiraceae and Ruminococcaceae [152] than those of healthy controls.
Another immune reaction caused by food, specifically to eating gluten, is celiac disease. In wheat grains, gluten is the key storage protein, constituting an intricate combination of hundreds of associated but dissimilar proteins, mainly glutenin and gliadin [153]. Rye, barley, and oats contain similar storage proteins, also collectively referred to as “gluten”, but with some differences from wheat gluten (for instance, oats appear to be tolerated by a majority of celiac individuals) [153]. Gluten consumption triggers the symptoms of celiac disease, which is an inherited genetic disorder and can only be treated by maintaining a strict gluten-free diet [154]. However, a significant diminution in beneficial gut bacteria has been observed as a result of consuming a gluten-free diet, raising concerns about the potential risks [155]. Hence, there is particular interest in probiotic supplements because they can balance the gut microbiome and provide the required nutrients to remain healthy [156,157,158]. Furthermore, certain bacteria in the gut are known to break down gluten protein, thereby increasing or decreasing its toxicity [159]. A number of studies have demonstrated that the human gastrointestinal tract, including the proximal small intestine, contains gluten-degrading bacteria such as Lactobacillus spp. or Rothia spp. [160,161,162,163,164]. Thus, celiac disease patients may benefit from increasing or decreasing certain types of bacteria in their gut in order to reduce gluten toxicity [159].

3.4. Impact of Early Feeding Choices on Allergy Development

Approximately two to five percent of children have food allergies that can cause mild to severe allergic reactions. As such, the immune system develops sequentially as a result of a series of coordinated and timed events beginning during pregnancy and continuing throughout the first few weeks of postpartum [165]. Due to the modulation of the immune system by dietary components, there is also an effect of early diet on allergic disease susceptibility [166,167]. A number of specific dietary components, such as milk, peanuts, eggs, and fish, are usual allergens. Infants who consume those components may experience a tough response of immune system. Additionally, dietary compounds, including tryptophan, vitamins A and D, omega-3 polyunsaturated fatty acids, dietary fiber, and acetic acid, interact with various receptors to regulate immune homeostasis, including G protein-coupled receptors and nuclear receptors [166,167].
Recent studies have found that the delayed introduction of solids does not appear to protect against allergic diseases. Moreover, the late introduction of solid foods could increase allergic sensitization risk to foods, inhalant allergens, and celiac disease in children [168]. The development of tolerance might be influenced by the interaction of the mucosal immune system with the allergen at the appropriate age; the protective effects may be improved by breastfeeding during the transition to weaning [168].
Numerous randomized clinical trials have demonstrated that early introduction of allergenic complementary foods before the age of 6 months, along with continued breastfeeding, can greatly decrease food allergy risks [169,170,171,172]. According to a recent European evidence-based guideline on allergy prevention, the most effective time to introduce peanuts with complementary feeding is between the ages of 4–6 months [171]. Furthermore, the randomized clinical trial evidence indicates that complementary feeding introduction at the age of 4–6 months did not result in a reduction in the rate and duration of subsequent breastfeeding [170,173]. Nevertheless, this relationship between complementary feeding practices and allergic diseases in children has produced conflicting results, and it is not completely clear at present [174]. In general terms, and according to updated guidelines for allergy prevention, there is a consensus on the delayed introduction of major allergenic foods and a greater risk of food allergies [174]. In this line, a number of studies have shown that early introduction of allergenic foods, such as eggs and peanuts, can diminish the risk of food allergies in the predisposed child population. There is, however, no evidence to suggest that this may be applicable to the entire child population [170]. For one child to be protected from food allergies, 63 children should be introduced to allergenic foods at an early age [170].

4. Microbiome and Food Intolerances

4.1. Understanding Food Intolerances

A food intolerance occurs when the digestive tract becomes inflamed or when the food cannot be digested properly due to non-immune reactions involving toxic, pharmacologic, and unknown mechanisms (for example, lack of digestive enzymes). Unlike food hypersensitivity, which is a disorder of the immune system caused by specific proteins in food [175,176]. The diagnosis of food intolerances can take a considerable amount of time. Although food intolerance is not a life-threatening condition, it may cause the sufferer to feel extremely unwell and can have a significant impact on their work and social lives [177]. It has been estimated that up to 15–20% of the population suffers from food intolerance, depending on definitions and data collection methods [178]. In fact, the estimation of food intolerances two decades ago was also around to 20% of the population [179]. There are many people with food intolerance who report gastrointestinal symptoms, and 50–84% of patients with functional gastrointestinal disorders, such as irritable bowel syndrome, perceive their symptoms to be related to food intolerance [180,181,182].
As a result of their heterogeneous pathophysiological mechanisms, non-immunological adverse reactions to food can be classified broadly as host-independent and host-dependent [183]. The food intolerances reported by patients with irritable bowel syndrome are an example of undefined food intolerances that cannot easily be explained by current pathophysiological mechanisms [184]. Generally, food intolerance manifests in more than one organ or system; however, gastrointestinal symptoms, such as abdominal pain, bloating, abdominal distension, diarrhea, and flatulence, are quite common. It is important to note that non-allergenic food reactions are dose-dependent, as opposed to food allergies, in which traces of food allergens can trigger severe reactions [184].

4.2. Food Intolerances and Microbiome

Components from dietary food that are undigested by host enzymes convert to bacterial substrates and are transformed into metabolites such as SCFAs that regulate gut homeostasis [185]. Various factors contribute to the development of food sensitivities, including the dysfunction of the intestinal barrier, which can be induced by gut microorganisms and pathogens [186]. There are fermentable carbohydrates naturally occurring in a wide variety of foods, such as fermentable polyols and oligo-di-mono-saccharides and polyols. Patients with functional gastrointestinal symptoms, such as those suffering from irritable bowel syndrome, have been identified to be sensitive to this group of carbohydrates [187,188].
It is important to note that food intolerance is attributed to a variety of non-immune pathways, such as lactose maldigestion, which is caused by a primary or secondary β-galactosidase deficiency (lactase). Due to this, undigested lactose moves to the colon, where it is fermented by the microbes from the gut, causing the production of gas (methane, carbon dioxide, and hydrogen) and bloating [159,189]. Adult human gut Bifidobacterium abundance is determined by genetic variations associated with lactose intolerance and dairy consumption [190]. There is evidence that certain gut symptoms experienced by lactose intolerance patients might be the result of an abundance of Bifidobacterium in the gut rather than a direct reaction to lactose consumption [191]. The findings of the aforementioned study [191] support the initial reports suggesting that lactose-fermenting bacteria could be responsible for lactose intolerance symptoms [192,193,194]. Moreover, in eight studies, Bifidobacterium administration and lactose tolerance were examined; most (5/8) reported a positive effect of Bifidobacterium on managing lactose intolerance symptoms [195]. Research on the efficacy of acute and chronic Bifidobacterium consumption is limited. In order to determine the time required to improve lactose digestion and tolerance using Bifidobacterium on a long-term basis, more studies are required [195].
In recent years, extensive research has been made into gut microbiome disruptions characterized by intestinal symptoms. One of those receiving growing interest is small intestinal bacterial overgrowth (SIBO) [196]. SIBO’s prevalence is increasing worldwide, with the most common risk factor being diet and the Western diet being considered unfavorable for this condition [197]. Different bacteria overgrowth has been predominately recognized in SIBO, such as Klebsiella pneumoniae, Escherichia coli, Prevotella, Streptococcus gramineus, etc. [196]. Furthermore, a recent study showed that bacteria enriched in SIBO patients appears to be correlated with constipation, with the Ruminococcaceae group defined as core bacteria in SIBO [198].

5. Practical Implications

5.1. Recommendations for Infant Feeding

Recommendations for infant feeding and its impact on the infant microbiome should be focused on (I) providing exclusive breastfeeding for the first 6 months, as breast milk contains beneficial bacteria that help establish a healthy gut microbiome and provides prebiotics, which are food for the beneficial bacteria in the infant’s gut [199]; (II) frequent skin-to-skin contact between mother and baby, which can be used to transfer beneficial bacteria from the mother’s skin to the baby, promoting a healthy microbiome [200]; (III) feeding on demand, which helps to maintain an optimal balance of gut bacteria and supports the natural development of the infant’s immune system [201]; (IV) avoiding the use of antibiotics for the infant unless absolutely necessary and prescribed by a healthcare professional, as they can disrupt the gut microbiome [202]; (V) a varied and balanced maternal diet that can positively influence the composition of breast milk and, consequently, the infant’s gut microbiome [203]; (VI) introducing a variety of solid foods to further support the development of a diverse microbiome around six months of age, and including foods that are rich in fiber and natural prebiotics to nourish the beneficial bacteria [199]; (VII) avoiding formula unless necessary, as it does not provide the same microbiome benefits as breast milk—if possible, minimize formula use and continue breastfeeding [204]; (VIII) under the guidance of a healthcare professional, probiotics and prebiotics supplements might be recommended for the infant to support a healthy gut microbiome [205,206,207], mainly based on probiotics like Lactobacillus rhamnosus, Bifidobacterium lactis, and Streptococcus thermophilus, or common prebiotics as galacto-oligosaccharides, fructo-oligosaccharides, and inulin [208]; and (IX) donor milk usage, if the mother’s milk is not available. Pasteurized donor breast milk is preferable to formula, as it still contains beneficial components that support microbiome development [209].

5.2. Breastfeeding Encouragement and Support

Many mothers face challenges that make breastfeeding difficult. Support and encouragement can help overcome these barriers. Informing parents about breastfeeding benefits and teaching proper techniques can enhance breastfeeding success [210], whereas healthcare providers, including lactation consultants and nurses, play a crucial role in offering support and addressing concerns [211]. Moreover, peer support groups provide emotional encouragement and practical advice from other breastfeeding mothers [212].
Creating breastfeeding-friendly workplaces with facilities for pumping and storing breast milk can help mothers continue breastfeeding after returning to work [213]. In this line, policies that promote breastfeeding, such as the Baby-Friendly Hospital Initiative, can support breastfeeding initiation and continuation [214]. Other important encouragement techniques should be focused on (I) encouraging words and positive feedback, which can boost a mother’s confidence in her ability to breastfeed [215]; (II) helping mothers address specific breastfeeding issues, such as latching difficulties or low milk supply, which can improve outcomes [216]; and (III) regular check-ins with healthcare providers or support groups, which can provide ongoing support and guidance [217].

5.3. Choosing the Right Formula Based on Its Components

The most common ingredients used in the formulation are proteins, fats, carbohydrates, vitamins, and minerals [218]. As a source of protein, whey protein isolates, skim milk powder, whey protein concentrate, among others, made from milk, are commonly used. The most utilized fat source is vegetable fat, although animal milk fats can also be used [219,220]. Infant formulas are generally regulated to ensure they provide the essential nutrients infants need [12]. Although fundamental formulations are essential, there are considerable variations between countries and regions [221]. The specific composition will depend on the age range (0–36 months) and the presence of any underlying health conditions or special nutritional requirements [12].
The majority of infant formulas are produced from cow’s milk [85], although alternative animals’ milk, including goat and sheep, have also been extensively marketed as potential substitutes for allergy management and due to their better lipid profile [85,222]. Another option is the use of formulas derived from soy proteins, which have been demonstrated to be effective for infants with galactosemia or congenital lactase deficiencies. These formulas have been shown to treat colic and milk allergies [85], but their use is not recommended for infants under 6 months due to the presence of various components, including phytoestrogens [85]. Protein hydrolysate formulas are designed for infants intolerant or allergic to cow’s milk or soy-based formulas [85,223].
In the present day, manufacturers consider rice proteins to be a valuable raw material due to their high nutritional profile and hypoallergenic properties. A rice grain’s protein content varies according to genotype, climate, and cultural practices but is typically between 7 and 10% [224,225]. Hydrolyzed rice protein formulas have been developed, and they appear to be a viable alternative to cow’s milk protein formulas, both for families who wish to feed their children plant-based meals and for children with cow’s milk protein allergies [226]. The use of hydrolyzed rice protein formulas has become increasingly popular in some countries for the feeding of infants who have allergies to cow’s milk proteins [227]. Due to the legal framework of foods for special medical purposes, both cow’s milk protein allergy formulas and the quality of rice proteins allow infants and children to be fed without the risk of nutritional deficiencies [228].
Amino acid formulas represent an alternative for infants with a severe allergy to cow’s milk who exhibit a reaction to or refusal to consume adequate amounts of hydrolysate formula [85]. Finally, there are specific formulas designed for infants with a congenital metabolic defect, low birth weight, or those with an exceptional medical or dietary condition [229]. For example, additives are employed in anti-regurgitation formulas to reduce acid reflux [229]; the exclusion of amino acids, such as phenylalanine, from infant formulas for infants with phenylketonuria [229]; and an additional example is the formulation of formulas with a higher protein/energy ratio for infants with low birth weight or premature birth [230].
Formulas also include novel ingredients such as HMOs that inhibit microorganism adhesion to the intestinal mucosa. This inhibits pathogens growth, the expression of inflammation genes, and the production of bacteriocins and organic acids [231]. An HMO is a complex carbohydrate, often called a targeted prebiotic [232,233,234,235]. Research indicates that HMOs play a significant role in infant immunity. HMOs have a unique structure that provides targeted benefits, such as enhancing the development of the gut barrier and balancing the immune system [232,233,234,235]. Infant formula with HMOs is safe, well tolerated, and supports the child’s growth at the appropriate age [236,237]. Secondary outcomes indicated that consumption of HMO-supplemented formula were associated with lower parent-reported morbidity (particularly bronchitis) and medication usage (antipyretics and antibiotics) [236]. Furthermore, HMOs support a healthy gut microbiome, as well as promote brain growth and cognitive function [237,238,239,240].
In line with WHO, ESPGHAN promotes optimal nutrition and health for infants and young children, emphasizing breastfeeding as well as complementary feeding that is appropriate and timely [241]. ESPGHAN considered that continued breastfeeding during the second year of life may be a desirable goal for some families and children, depending on individual factors, personal preferences, and environmental factors [241]. It is recommended that complementary foods are introduced at a suitable age: Exclusive or full breastfeeding should be promoted for at least 4 months, and predominant/exclusive breastfeeding for 6 months is considered an appropriate goal. If a child or infant has an allergy to food, complementary foods with high allergenic potential (e.g., cooked egg, peanuts, or eggs) should be introduced in an age-appropriate manner when supplementation first begins at 4 months (17 weeks) [241].

5.4. Guidelines for Introducing Solid Foods

The introduction of solid foods represents a crucial element in the maintenance of optimal nutritional status, the prevention of potential allergic reactions, the reduction of the risk of disease, and the improvement of the modulation of the gut microbiome. Different aspects must be considered during the introduction of complementary foods, which typically occurs between 6 and 23 months of age, as this period is crucial for physical and cognitive development [242]:
(a) Choosing the adequate moment for every infant: In accordance with the recommendations of the AAP, WHO, and ESPGHAN, the introduction of solid foods is advised to commence between 4 and 6 months, based on the child’s developmental stage (e.g., the ability to sit in a chair and hold their heads up, interest in food, opening their mouths when approached with a spoon of food, putting their hands or toys in their mouths, loss of the tongue thrust reflex, etc.) [242,243]. It must be noted that an early introduction of solid foods may have negative effects. For instance, if the digestive and immune systems are not sufficiently developed, there are higher probabilities of choking, acute gastroenteritis, and upper respiratory tract infections [244].
(b) Food texture/consistency: Initial foods should be pureed, mashed, or semi-solid. At the age of 8–12 months, the introduction of foods that can be consumed with the hands is recommended. Ultimately, after 12 months of age, most children will have reached the same dietary texture options as the rest of the family [35,245]. During this period, the modification and exposure to a wide variety of textures is recommended to encourage the acceptance and enjoyment of food [93].
(c) Food quantity: The quantity, as well as consistency, should be increased gradually in accordance with the child’s growth and development [242]. However, the appropriate number of meals is dependent on the energy density of the food and the typical quantities consumed at each meal, which is typically two to three times a day until three to four times a day after 12 months [242].
(d) Food quality: Initial foods must be rich in iron and other minerals, such as zinc [35]. In this context, meat such as chicken or pork, eggs, or enriched cereals that can be mixed with breast milk, including oatmeal or rice, are recommended [242].
(e) Introduction of new foods and increasing acceptance: It is recommended that foods be introduced one at a time, with a recommended interval of 3 to 5 days between each introduction [35]. Due to the potential for allergic reactions, it is important to monitor for any adverse food-related responses. Extant evidence suggests that the early introduction of potentially allergenic foods, at approximately six months of age, may result in a gradual reduction in the risk of food allergy [93]. For enhancing tolerance to specific flavors, repeated exposures over a period of time between six and thirty six times are recommended [242]. The introduction of foods containing gluten, processed meats, fish, and shellfish should be carried out with caution due to the potential presence of components that may pose a significant health risk for the child [246,247].
(f) Some examples of appropriate first foods: soft pieces of banana, scrambled eggs, well-cooked pasta, well-cooked or mashed potatoes or peas, etc. They may help to prevent choking [248].

5.5. Future Research Directions

Food allergies and intolerances are more common in children than in other age groups. While food intolerances are more prevalent and less harmful, food allergies can be life-threatening. Therefore, a better understanding of the adverse effects of foods on functional disorders is needed [249]. Moreover, the dramatic global increase in the prevalence of these disorders requires effective strategies, considering the environmental factors, with emphasis on the study of the unknown underlying mechanisms, especially those related to the microbiome and immune system [126].
Although breastfeeding is the most recommended feeding method in infancy, some aspects, like the presence of allergens in breast milk or the effect of early introduction of allergenic foods, are still inconclusive. Thus, possible modifying factors (e.g., HMO and SCFA composition, the introduction of solid food, and immunologic factors) should be studied more in depth [250]. In this line, it has been hypothesized that regulating the composition of breastmilk, including the microbiome, could prevent allergic diseases early in life [251]. On the other hand, diet-derived metabolites determine the biologic effects of diet, and they vary according to different metabolic pathways in the host and in the gut microbiome, which may contribute to the heterogeneity observed when evaluating responses to diet interventions [252].
Microbiome research needs to be improved with appropriate experimental, methodological, and statistical designs. In this line, some recommendations have been made, including (I) selecting the adequate sampling frequencies, (II) using a combination of available methods in microbiome studies, (III) considering interactions between microorganisms and their hosts, or (IV) implementing a toolbox of technologies and more cultivation-based approaches into microbiome research [253]. Furthermore, different factors, such as the use of antibiotics or breastfeeding, must be considered in all trial designs, especially in those with longer duration and follow-up periods [254].
Although the functional understanding of the microbiome has grown considerably with the addition of metabolomics, metatranscriptomics and metaproteomics, and metagenomics, effort is still needed to develop new ex vivo assays targeting panels of entire microbiomes, simple microbial communities, or individual bacterium [255]. Furthermore, a computational approach integrating multiple data layers instead of evaluating individual data would allow the validation of mechanistic insights and new discoveries [256].
Based on the above, longitudinal studies can help elucidate the mechanisms through which the microbiome influences immune responses and the development of allergies. They can reveal how different feeding practices affect the microbiome’s composition and function over time. Moreover, results from these studies can inform guidelines on infant feeding practices, emphasizing the importance of breastfeeding for microbiome health. They can also lead to the development of probiotics or dietary supplements aimed at mimicking the beneficial effects of breastfeeding for formula-fed infants. Finally, findings from longitudinal research can shape public health policies and initiatives aimed at promoting breastfeeding and supporting mothers in breastfeeding practices [257,258].

6. Conclusions

Breastfeeding continues to be the most recommended method for infant feeding, with particular emphasis during the first six months of life, as it is associated with a gut microbiome rich in microorganisms that digest human milk. Introducing solid foods into an infant’s diet modifies this microbiome, evolving from a simple environment to one that is more complex. The early introduction (before the age of 6 months) of allergenic complementary foods, along with continued breastfeeding, could greatly reduce the risk of allergies and food intolerances. Individuals with food intolerances and allergies present different gut microbiome patterns, which could be both the cause and consequence of several related complications. Further research is needed to fully understand the role of the microbiome, the functional interactions, and its implication in food allergies and intolerance pathogenesis.

Author Contributions

Conceptualization, L.H.-Q. and J.P.-D.; methodology, L.H.-Q. and H.V.-L.; software, J.P.-D.; validation, J.P.-D.; writing—original draft preparation, J.P.-D., L.H.-Q., H.V.-L. and D.H.-N.; writing—review and editing, D.H.-N. and J.P.-D.; visualization, L.H.-Q. and J.P.-D.; supervision J.P.-D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Julio Plaza-Diaz is part of the “UGR Plan Propio de Investigación 2016” and the “Excellence actions: Unit of Excellence on Exercise and Health (UCEES), University of Granada”.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Berti, C.; Socha, P. Infant and Young Child Feeding Practices and Health. Nutrients 2023, 15, 1184. [Google Scholar] [CrossRef]
  2. World Health Organization (WHO). Infant and Young Child Feeding. Available online: https://www.who.int/news-room/fact-sheets/detail/infant-and-young-child-feeding (accessed on 24 July 2024).
  3. Rollins, N.C.; Bhandari, N.; Hajeebhoy, N.; Horton, S.; Lutter, C.K.; Martines, J.C.; Piwoz, E.G.; Richter, L.M.; Victora, C.G. Why invest, and what it will take to improve breastfeeding practices? Lancet 2016, 387, 491–504. [Google Scholar] [CrossRef]
  4. Laxmi, A.; Amanda, L.; Dolly, D.; Rubina, G. Infant Feeding Practices: A Global Perspective; IntechOpen: London, UK, 2023. [Google Scholar]
  5. Hirani, S.A.A.; Richter, S.; Salami, B.; Vallianatos, H. Sociocultural Factors Affecting Breastfeeding Practices of Mothers During Natural Disasters: A Critical Ethnography in Rural Pakistan. Glob. Qual. Nurs. Res. 2023, 10, 23333936221148808. [Google Scholar] [CrossRef]
  6. Reinsma, K.; Bolima, N.; Fonteh, F.; Okwen, P.; Yota, D.; Montgomery, S. Incorporating cultural beliefs in promoting exclusive breastfeeding. Afr. J. Midwifery Womens Health 2012, 6, 65–70. [Google Scholar] [CrossRef]
  7. UNICEF. Infant and Young Child Feeding. Available online: https://data.unicef.org/topic/nutrition/infant-and-young-child-feeding/ (accessed on 24 July 2024).
  8. World Health Organization (WHO). Breastfeeding. Available online: https://www.who.int/health-topics/breastfeeding#tab=tab_1 (accessed on 24 July 2024).
  9. World Health Organization (WHO). Exclusively Breastfeed for 6 Months. Available online: https://www.emro.who.int/nutrition/breastfeeding/index.html (accessed on 25 July 2024).
  10. Pan American Health Organization (PAHO). Exclusive Breastfeeding in Infant under Six Months of Age. Available online: https://www.paho.org/en/enlace/exclusive-breastfeeding-infant-under-six-months-age (accessed on 25 July 2024).
  11. Yan, J.; Liu, L.; Zhu, Y.; Huang, G.; Wang, P.P. The association between breastfeeding and childhood obesity: A meta-analysis. BMC Public Health 2014, 14, 1267. [Google Scholar] [CrossRef] [PubMed]
  12. Bakshi, S.; Paswan, V.K.; Yadav, S.P.; Bhinchhar, B.K.; Kharkwal, S.; Rose, H.; Kanetkar, P.; Kumar, V.; Al-Zamani, Z.A.S.; Bunkar, D.S. A comprehensive review on infant formula: Nutritional and functional constituents, recent trends in processing and its impact on infants’ gut microbiota. Front. Nutr. 2023, 10, 1194679. [Google Scholar] [CrossRef] [PubMed]
  13. National Academies of Sciences, Engineering, and Medicine. Feeding Infants and Children from Birth to 24 Months: Summarizing Existing Guidance; National Academies Press: Washington, DC, USA, 2020. [Google Scholar]
  14. Health Canada; Canadian Paediatric Society; Dietitians of Canada; Breastfeeding Committee for Canada. Nutrition for healthy term infants: Recommendations from birth to six months. Can. J. Diet. Pract. Res. 2012, 73, 204. [Google Scholar] [CrossRef] [PubMed]
  15. Schwartz, C.; Scholtens, P.A.; Lalanne, A.; Weenen, H.; Nicklaus, S. Development of healthy eating habits early in life. Review of recent evidence and selected guidelines. Appetite 2011, 57, 796–807. [Google Scholar] [CrossRef] [PubMed]
  16. Klerks, M.; Roman, S.; Bernal, M.J.; Haro-Vicente, J.F.; Sanchez-Siles, L.M. Complementary Feeding Practices and Parental Pressure to Eat among Spanish Infants and Toddlers: A Cross-Sectional Study. Int. J. Environ. Res. Public Health 2021, 18, 1982. [Google Scholar] [CrossRef]
  17. Low, F.M.; Gluckman, P.D.; Hanson, M.A. Maternal and child health: Is making ‘healthy choices’ an oxymoron? Glob. Health Promot. 2021, 28, 66–69. [Google Scholar] [CrossRef]
  18. Maier-Noth, A.; Schaal, B.; Leathwood, P.; Issanchou, S. The Lasting Influences of Early Food-Related Variety Experience: A Longitudinal Study of Vegetable Acceptance from 5 Months to 6 Years in Two Populations. PLoS ONE 2016, 11, e0151356. [Google Scholar] [CrossRef] [PubMed]
  19. Nicklaus, S. The role of food experiences during early childhood in food pleasure learning. Appetite 2016, 104, 3–9. [Google Scholar] [CrossRef] [PubMed]
  20. Faber, M.; Malan, L.; Kruger, H.S.; Asare, H.; Visser, M.; Mukwevho, T.; Ricci, C.; Smuts, C.M. Potential of Egg as Complementary Food to Improve Nutrient Intake and Dietary Diversity. Nutrients 2022, 14, 3396. [Google Scholar] [CrossRef]
  21. Huang, H.; Gao, Y.; Zhu, N.; Yuan, G.; Li, X.; Feng, Y.; Gao, L.; Yu, J. The Effects of Breastfeeding for Four Months on Thinness, Overweight, and Obesity in Children Aged 3 to 6 Years: A Retrospective Cohort Study from National Physical Fitness Surveillance of Jiangsu Province, China. Nutrients 2022, 14, 4154. [Google Scholar] [CrossRef]
  22. Gillman, M.W.; Ludwig, D.S. How early should obesity prevention start? N. Engl. J. Med. 2013, 369, 2173–2175. [Google Scholar] [CrossRef]
  23. Birch, L.L.; Doub, A.E. Learning to eat: Birth to age 2 y. Am. J. Clin. Nutr. 2014, 99, 723S–728S. [Google Scholar] [CrossRef] [PubMed]
  24. Hermoso, M.; Tabacchi, G.; Iglesia-Altaba, I.; Bel-Serrat, S.; Moreno-Aznar, L.A.; García-Santos, Y.; García-Luzardo, M.d.R.; Santana-Salguero, B.; Peña-Quintana, L.; Serra-Majem, L. The nutritional requirements of infants. Towards EU alignment of reference values: The EURRECA network. Matern. Child Nutr. 2010, 6, 55–83. [Google Scholar] [CrossRef]
  25. Pearce, J.; Taylor, M.; Langley-Evans, S. Timing of the introduction of complementary feeding and risk of childhood obesity: A systematic review. Int. J. Obes. 2013, 37, 1295–1306. [Google Scholar] [CrossRef]
  26. Sun, C.; Foskey, R.J.; Allen, K.J.; Dharmage, S.C.; Koplin, J.J.; Ponsonby, A.-L.; Lowe, A.J.; Matheson, M.C.; Tang, M.L.; Gurrin, L. The impact of timing of introduction of solids on infant body mass index. J. Pediatr. 2016, 179, 104–110.e1. [Google Scholar] [CrossRef]
  27. Brophy, S.; Cooksey, R.; Gravenor, M.B.; Mistry, R.; Thomas, N.; Lyons, R.A.; Williams, R. Risk factors for childhood obesity at age 5: Analysis of the millennium cohort study. BMC Public Health 2009, 9, 467. [Google Scholar] [CrossRef]
  28. Huh, S.Y.; Rifas-Shiman, S.L.; Taveras, E.M.; Oken, E.; Gillman, M.W. Timing of solid food introduction and risk of obesity in preschool-aged children. Pediatrics 2011, 127, e544–e551. [Google Scholar] [CrossRef] [PubMed]
  29. Zheng, J.-S.; Liu, H.; Zhao, Y.-M.; Li, J.; Chen, Y.; Zhu, S.; Chen, H.; Huang, T.; Li, D. Complementary feeding and childhood adiposity in preschool-aged children in a large Chinese cohort. J. Pediatr. 2015, 166, 326–331.e2. [Google Scholar] [CrossRef] [PubMed]
  30. Gartner, L.M.; Morton, J.; Lawrence, R.A.; Naylor, A.J.; O’Hare, D.; Schanler, R.J.; Eidelman, A.I. Breastfeeding and the use of human milk. Pediatrics 2005, 115, 496–506. [Google Scholar]
  31. Critch, J.N.; Canadian Paediatric Society, Nutrition and Gastroenterology Committee. Nutrition for healthy term infants, birth to six months: An overview. Paediatr. Child. Health 2013, 18, 206–207. [Google Scholar] [PubMed]
  32. Boland, M. Exclusive breastfeeding should continue to six months. Paediatr. Child Health 2005, 10, 148. [Google Scholar] [CrossRef]
  33. D’Hollander, C.J.; Keown-Stoneman, C.D.; Birken, C.S.; O’Connor, D.L.; Maguire, J.L.; Cohn, R.; Lau, E.; Laupacis, A.; Parkin, P.C.; Salter, M. Timing of introduction to solid food, growth, and nutrition risk in later childhood. J. Pediatr. 2022, 240, 102–109.e3. [Google Scholar] [CrossRef]
  34. Kramer, M.S.; Kakuma, R. Optimal duration of exclusive breastfeeding. Cochrane Database Syst. Rev. 2012, 2012, CD003517. [Google Scholar] [CrossRef] [PubMed]
  35. Fewtrell, M.; Bronsky, J.; Campoy, C.; Domellöf, M.; Embleton, N.; Mis, N.F.; Hojsak, I.; Hulst, J.M.; Indrio, F.; Lapillonne, A. Complementary feeding: A position paper by the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) Committee on Nutrition. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 119–132. [Google Scholar] [CrossRef]
  36. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Vilchez-Padial, L.M.; Gil, A. Evidence of the Anti-Inflammatory Effects of Probiotics and Synbiotics in Intestinal Chronic Diseases. Nutrients 2017, 9, 555. [Google Scholar] [CrossRef]
  37. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Immune-Mediated Mechanisms of Action of Probiotics and Synbiotics in Treating Pediatric Intestinal Diseases. Nutrients 2018, 10, 42. [Google Scholar] [CrossRef]
  38. Plaza-Diaz, J.; Gomez-Fernandez, A.; Chueca, N.; Torre-Aguilar, M.J.; Gil, A.; Perez-Navero, J.L.; Flores-Rojas, K.; Martin-Borreguero, P.; Solis-Urra, P.; Ruiz-Ojeda, F.J.; et al. Autism Spectrum Disorder (ASD) with and without Mental Regression is Associated with Changes in the Fecal Microbiota. Nutrients 2019, 11, 337. [Google Scholar] [CrossRef] [PubMed]
  39. Alvarez-Mercado, A.I.; Navarro-Oliveros, M.; Robles-Sanchez, C.; Plaza-Diaz, J.; Saez-Lara, M.J.; Munoz-Quezada, S.; Fontana, L.; Abadia-Molina, F. Microbial Population Changes and Their Relationship with Human Health and Disease. Microorganisms 2019, 7, 68. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, H.; Sparks, J.B.; Karyala, S.V.; Settlage, R.; Luo, X.M. Host adaptive immunity alters gut microbiota. ISME J. 2015, 9, 770–781. [Google Scholar] [CrossRef]
  41. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
  42. Hakansson, A.; Molin, G. Gut microbiota and inflammation. Nutrients 2011, 3, 637–682. [Google Scholar] [CrossRef] [PubMed]
  43. Collins, S.L.; Patterson, A.D. The gut microbiome: An orchestrator of xenobiotic metabolism. Acta Pharm. Sin. B 2020, 10, 19–32. [Google Scholar] [CrossRef]
  44. Wong, J.M.; De Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  45. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef]
  46. Wen, L.; Duffy, A. Factors Influencing the Gut Microbiota, Inflammation, and Type 2 Diabetes. J. Nutr. 2017, 147, 1468S–1475S. [Google Scholar] [CrossRef]
  47. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  48. Kumbhare, S.V.; Patangia, D.V.; Patil, R.H.; Shouche, Y.S.; Patil, N.P. Factors influencing the gut microbiome in children: From infancy to childhood. J. Biosci. 2019, 44, 49. [Google Scholar] [CrossRef]
  49. Raspini, B.; Vacca, M.; Porri, D.; De Giuseppe, R.; Calabrese, F.M.; Chieppa, M.; Liso, M.; Cerbo, R.M.; Civardi, E.; Garofoli, F.; et al. Early Life Microbiota Colonization at Six Months of Age: A Transitional Time Point. Front. Cell Infect. Microbiol. 2021, 11, 590202. [Google Scholar] [CrossRef] [PubMed]
  50. Bogaert, D.; van Beveren, G.J.; de Koff, E.M.; Lusarreta Parga, P.; Balcazar Lopez, C.E.; Koppensteiner, L.; Clerc, M.; Hasrat, R.; Arp, K.; Chu, M.; et al. Mother-to-infant microbiota transmission and infant microbiota development across multiple body sites. Cell Host Microbe 2023, 31, 447–460.e6. [Google Scholar] [CrossRef] [PubMed]
  51. Thompson, A.L.; Monteagudo-Mera, A.; Cadenas, M.B.; Lampl, M.L.; Azcarate-Peril, M.A. Milk-and solid-feeding practices and daycare attendance are associated with differences in bacterial diversity, predominant communities, and metabolic and immune function of the infant gut microbiome. Front. Cell. Infect. Microbiol. 2015, 5, 3. [Google Scholar] [CrossRef] [PubMed]
  52. Stewart, C.J.; Ajami, N.J.; O’Brien, J.L.; Hutchinson, D.S.; Smith, D.P.; Wong, M.C.; Ross, M.C.; Lloyd, R.E.; Doddapaneni, H.; Metcalf, G.A. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 2018, 562, 583–588. [Google Scholar] [CrossRef]
  53. Moore, R.E.; Townsend, S.D. Temporal development of the infant gut microbiome. Open Biol. 2019, 9, 190128. [Google Scholar] [CrossRef]
  54. Leyva, L.L.; Brereton, N.J.; Koski, K.G. Emerging frontiers in human milk microbiome research and suggested primers for 16S rRNA gene analysis. Comput. Struct. Biotechnol. J. 2021, 19, 121–133. [Google Scholar] [CrossRef]
  55. Kim, S.Y.; Yi, D.Y. Analysis of the human breast milk microbiome and bacterial extracellular vesicles in healthy mothers. Exp. Mol. Med. 2020, 52, 1288–1297. [Google Scholar] [CrossRef]
  56. Moubareck, C.A. Human milk microbiota and oligosaccharides: A glimpse into benefits, diversity, and correlations. Nutrients 2021, 13, 1123. [Google Scholar] [CrossRef]
  57. Demmelmair, H.; Jiménez, E.; Collado, M.C.; Salminen, S.; McGuire, M.K. Maternal and perinatal factors associated with the human milk microbiome. Curr. Dev. Nutr. 2020, 4, nzaa027. [Google Scholar]
  58. Enaud, R.; Prevel, R.; Ciarlo, E.; Beaufils, F.; Wieërs, G.; Guery, B.; Delhaes, L. The gut-lung axis in health and respiratory diseases: A place for inter-organ and inter-kingdom crosstalks. Front. Cell. Infect. Microbiol. 2020, 10, 9. [Google Scholar] [CrossRef] [PubMed]
  59. Sánchez, C.; Franco, L.; Regal, P.; Lamas, A.; Cepeda, A.; Fente, C. Breast milk: A source of functional compounds with potential application in nutrition and therapy. Nutrients 2021, 13, 1026. [Google Scholar] [CrossRef]
  60. Oikonomou, G.; Addis, M.F.; Chassard, C.; Nader-Macias, M.E.F.; Grant, I.; Delbès, C.; Bogni, C.I.; Le Loir, Y.; Even, S. Milk microbiota: What are we exactly talking about? Front. Microbiol. 2020, 11, 60. [Google Scholar] [CrossRef] [PubMed]
  61. Bokulich, N.A.; Chung, J.; Battaglia, T.; Henderson, N.; Jay, M.; Li, H.; Lieber, A.D.; Wu, F.; Perez-Perez, G.I.; Chen, Y. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016, 8, 343ra82. [Google Scholar] [CrossRef] [PubMed]
  62. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef]
  63. Shao, Y.; Forster, S.C.; Tsaliki, E.; Vervier, K.; Strang, A.; Simpson, N.; Kumar, N.; Stares, M.D.; Rodger, A.; Brocklehurst, P. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature 2019, 574, 117–121. [Google Scholar] [CrossRef]
  64. Reyman, M.; van Houten, M.A.; van Baarle, D.; Bosch, A.A.; Man, W.H.; Chu, M.L.J.; Arp, K.; Watson, R.L.; Sanders, E.A.; Fuentes, S. Impact of delivery mode-associated gut microbiota dynamics on health in the first year of life. Nat. Commun. 2019, 10, 4997. [Google Scholar] [CrossRef]
  65. 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]
  66. Derrien, M.; Alvarez, A.S.; de Vos, W.M. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019, 27, 997–1010. [Google Scholar] [CrossRef]
  67. Huang, H.; Jiang, J.; Wang, X.; Jiang, K.; Cao, H. Exposure to prescribed medication in early life and impacts on gut microbiota and disease development. EClinicalMedicine 2024, 68, 102428. [Google Scholar] [CrossRef]
  68. van den Elsen, L.W.J.; Garssen, J.; Burcelin, R.; Verhasselt, V. Shaping the Gut Microbiota by Breastfeeding: The Gateway to Allergy Prevention? Front. Pediatr. 2019, 7, 47. [Google Scholar] [CrossRef]
  69. Pannaraj, P.S.; Li, F.; Cerini, C.; Bender, J.M.; Yang, S.; Rollie, A.; Adisetiyo, H.; Zabih, S.; Lincez, P.J.; Bittinger, K. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017, 171, 647–654. [Google Scholar] [CrossRef] [PubMed]
  70. Roger, L.C.; Costabile, A.; Holland, D.T.; Hoyles, L.; McCartney, A.L. Examination of faecal Bifidobacterium populations in breast-and formula-fed infants during the first 18 months of life. Microbiology 2010, 156, 3329–3341. [Google Scholar] [CrossRef]
  71. Roger, L.C.; McCartney, A.L. Longitudinal investigation of the faecal microbiota of healthy full-term infants using fluorescence in situ hybridization and denaturing gradient gel electrophoresis. Microbiology 2010, 156, 3317–3328. [Google Scholar] [CrossRef] [PubMed]
  72. Bridgman, S.L.; Azad, M.B.; Field, C.J.; Haqq, A.M.; Becker, A.B.; Mandhane, P.J.; Subbarao, P.; Turvey, S.E.; Sears, M.R.; Scott, J.A. Fecal short-chain fatty acid variations by breastfeeding status in infants at 4 months: Differences in relative versus absolute concentrations. Front. Nutr. 2017, 4, 11. [Google Scholar] [CrossRef]
  73. Ma, J.; Li, Z.; Zhang, W.; Zhang, C.; Zhang, Y.; Mei, H.; Zhuo, N.; Wang, H.; Wang, L.; Wu, D. Comparison of gut microbiota in exclusively breast-fed and formula-fed babies: A study of 91 term infants. Sci. Rep. 2020, 10, 15792. [Google Scholar] [CrossRef] [PubMed]
  74. Walker, W.A.; Iyengar, R.S. Breast milk, microbiota, and intestinal immune homeostasis. Pediatr. Res. 2015, 77, 220–228. [Google Scholar] [CrossRef]
  75. Levin, A.M.; Sitarik, A.R.; Havstad, S.L.; Fujimura, K.E.; Wegienka, G.; Cassidy-Bushrow, A.E.; Kim, H.; Zoratti, E.M.; Lukacs, N.W.; Boushey, H.A. Joint effects of pregnancy, sociocultural, and environmental factors on early life gut microbiome structure and diversity. Sci. Rep. 2016, 6, 31775. [Google Scholar] [CrossRef] [PubMed]
  76. Donald, K.; Petersen, C.; Turvey, S.E.; Finlay, B.B.; Azad, M.B. Secretory IgA: Linking microbes, maternal health, and infant health through human milk. Cell Host Microbe 2022, 30, 650–659. [Google Scholar] [CrossRef]
  77. Dzidic, M.; Mira, A.; Artacho, A.; Abrahamsson, T.R.; Jenmalm, M.C.; Collado, M.C. Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life. Pediatr. Allergy Immunol. 2020, 31, 250–257. [Google Scholar] [CrossRef]
  78. Orndorff, P.E.; Devapali, A.; Palestrant, S.; Wyse, A.; Everett, M.L.; Bollinger, R.R.; Parker, W. Immunoglobulin-mediated agglutination of and biofilm formation by Escherichia coli K-12 require the type 1 pilus fiber. Infect. Immun. 2004, 72, 1929–1938. [Google Scholar] [CrossRef]
  79. Pannaraj, P.S.; Ly, M.; Cerini, C.; Saavedra, M.; Aldrovandi, G.M.; Saboory, A.A.; Johnson, K.M.; Pride, D.T. Shared and distinct features of human milk and infant stool viromes. Front. Microbiol. 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed]
  80. Milani, C.; Casey, E.; Lugli, G.A.; Moore, R.; Kaczorowska, J.; Feehily, C.; Mangifesta, M.; Mancabelli, L.; Duranti, S.; Turroni, F. Tracing mother-infant transmission of bacteriophages by means of a novel analytical tool for shotgun metagenomic datasets: METAnnotatorX. Microbiome 2018, 6, 145. [Google Scholar] [CrossRef]
  81. Davis, E.C.; Castagna, V.P.; Sela, D.A.; Hillard, M.A.; Lindberg, S.; Mantis, N.J.; Seppo, A.E.; Jarvinen, K.M. Gut microbiome and breast-feeding: Implications for early immune development. J. Allergy Clin. Immunol. 2022, 150, 523–534. [Google Scholar] [CrossRef] [PubMed]
  82. da Silva, R.C.; Ferdaus, M.J. Technological Advances in Infant Formula Ingredients; IntechOpen: London, UK, 2023. [Google Scholar]
  83. de Almagro Garcia, M.C.; JA, M.M. New ingredients in infant formula. Health and functional benefits. Nutr. Hosp. 2017, 34, 8–12. [Google Scholar] [PubMed]
  84. Hedrick, J.; Yeiser, M.; Harris, C.L.; Wampler, J.L.; London, H.E.; Patterson, A.C.; Wu, S.S. Infant formula with added bovine milk fat globule membrane and modified iron supports growth and normal iron status at one year of age: A randomized controlled trial. Nutrients 2021, 13, 4541. [Google Scholar] [CrossRef]
  85. Martin, C.R.; Ling, P.R.; Blackburn, G.L. Review of Infant Feeding: Key Features of Breast Milk and Infant Formula. Nutrients 2016, 8, 279. [Google Scholar] [CrossRef]
  86. Ferreres-Serafini, L.; Martin-Orue, S.M.; Sadurni, M.; Jimenez, J.; Moreno-Munoz, J.A.; Castillejos, L. Supplementing infant milk formula with a multi-strain synbiotic and osteopontin enhances colonic microbial colonization and modifies jejunal gene expression in lactating piglets. Food Funct. 2024, 15, 6536–6552. [Google Scholar] [CrossRef] [PubMed]
  87. Chouraqui, J.P.; Grathwohl, D.; Labaune, J.M.; Hascoet, J.M.; de Montgolfier, I.; Leclaire, M.; Giarre, M.; Steenhout, P. Assessment of the safety, tolerance, and protective effect against diarrhea of infant formulas containing mixtures of probiotics or probiotics and prebiotics in a randomized controlled trial. Am. J. Clin. Nutr. 2008, 87, 1365–1373. [Google Scholar] [CrossRef]
  88. Braegger, C.; Chmielewska, A.; Decsi, T.; Kolacek, S.; Mihatsch, W.; Moreno, L.; Piescik, M.; Puntis, J.; Shamir, R.; Szajewska, H.; et al. Supplementation of infant formula with probiotics and/or prebiotics: A systematic review and comment by the ESPGHAN committee on nutrition. J. Pediatr. Gastroenterol. Nutr. 2011, 52, 238–250. [Google Scholar] [CrossRef]
  89. Fallani, M.; Amarri, S.; Uusijarvi, A.; Adam, R.; Khanna, S.; Aguilera, M.; Gil, A.; Vieites, J.M.; Norin, E.; Young, D. Determinants of the human infant intestinal microbiota after the introduction of first complementary foods in infant samples from five European centres. Microbiology 2011, 157, 1385–1392. [Google Scholar] [CrossRef] [PubMed]
  90. Zimmer, J.; Lange, B.; Frick, J.-S.; Sauer, H.; Zimmermann, K.; Schwiertz, A.; Rusch, K.; Klosterhalfen, S.; Enck, P. A vegan or vegetarian diet substantially alters the human colonic faecal microbiota. Eur. J. Clin. Nutr. 2012, 66, 53–60. [Google Scholar] [CrossRef]
  91. Johnson, C.L.; Versalovic, J. The human microbiome and its potential importance to pediatrics. Pediatrics 2012, 129, 950–960. [Google Scholar] [CrossRef] [PubMed]
  92. Ku, H.J.; Kim, Y.T.; Lee, J.H. Microbiome Study of Initial Gut Microbiota from Newborn Infants to Children Reveals that Diet Determines Its Compositional Development. J. Microbiol. Biotechnol. 2020, 30, 1067–1071. [Google Scholar] [CrossRef] [PubMed]
  93. Borowitz, S.M. First Bites-Why, When, and What Solid Foods to Feed Infants. Front. Pediatr. 2021, 9, 654171. [Google Scholar] [CrossRef]
  94. Homann, C.M.; Rossel, C.A.J.; Dizzell, S.; Bervoets, L.; Simioni, J.; Li, J.; Gunn, E.; Surette, M.G.; de Souza, R.J.; Mommers, M.; et al. Infants’ First Solid Foods: Impact on Gut Microbiota Development in Two Intercontinental Cohorts. Nutrients 2021, 13, 2639. [Google Scholar] [CrossRef]
  95. 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. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  96. Arrieta, M.-C.; Stiemsma, L.T.; Amenyogbe, N.; Brown, E.M.; Finlay, B. The intestinal microbiome in early life: Health and disease. Front. Immunol. 2014, 5, 427. [Google Scholar] [CrossRef]
  97. Li, Z.; Zhou, J.; Liang, H.; Ye, L.; Lan, L.; Lu, F.; Wang, Q.; Lei, T.; Yang, X.; Cui, P.; et al. Differences in Alpha Diversity of Gut Microbiota in Neurological Diseases. Front. Neurosci. 2022, 16, 879318. [Google Scholar] [CrossRef]
  98. Wang, J.; Zhou, Y.; Zhang, H.; Hu, L.; Liu, J.; Wang, L.; Wang, T.; Zhang, H.; Cong, L.; Wang, Q. Pathogenesis of allergic diseases and implications for therapeutic interventions. Signal Transduct. Target. Ther. 2023, 8, 138. [Google Scholar] [CrossRef]
  99. Wanka, L.; Jappe, U. Trained immunity and allergy: State of the art and future perspectives. Allergy 2021, 76, 1265–1267. [Google Scholar] [CrossRef] [PubMed]
  100. Fujimura, K.E.; Lynch, S.V. Microbiota in allergy and asthma and the emerging relationship with the gut microbiome. Cell Host Microbe 2015, 17, 592–602. [Google Scholar] [CrossRef] [PubMed]
  101. Stark, J.F. Bacteriology in the service of sanitation: The factory environment and the regulation of industrial anthrax in late-victorian Britain. Social Hist. Med. 2012, 25, 343–361. [Google Scholar] [CrossRef]
  102. Zhao, H.; Liao, X.; Kang, Y. Tregs: Where we are and what comes next? Front. Immunol. 2017, 8, 1578. [Google Scholar] [CrossRef]
  103. Chaudhary, B.; Elkord, E. Regulatory T cells in the tumor microenvironment and cancer progression: Role and therapeutic targeting. Vaccines 2016, 4, 28. [Google Scholar] [CrossRef]
  104. Miyara, M.; Yoshioka, Y.; Kitoh, A.; Shima, T.; Wing, K.; Niwa, A.; Parizot, C.; Taflin, C.; Heike, T.; Valeyre, D. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009, 30, 899–911. [Google Scholar] [CrossRef]
  105. Duhen, T.; Duhen, R.; Lanzavecchia, A.; Sallusto, F.; Campbell, D.J. Functionally distinct subsets of human FOXP3+ Treg cells that phenotypically mirror effector Th cells. Blood J. Am. Soc. Hematol. 2012, 119, 4430–4440. [Google Scholar] [CrossRef]
  106. Dong, S.; Maiella, S.; Xhaard, A.; Pang, Y.; Wenandy, L.; Larghero, J.; Becavin, C.; Benecke, A.; Bianchi, E.; Socié, G. Multiparameter single-cell profiling of human CD4+ FOXP3+ regulatory T-cell populations in homeostatic conditions and during graft-versus-host disease. Blood J. Am. Soc. Hematol. 2013, 122, 1802–1812. [Google Scholar] [CrossRef]
  107. Mason, G.M.; Lowe, K.; Melchiotti, R.; Ellis, R.; de Rinaldis, E.; Peakman, M.; Heck, S.; Lombardi, G.; Tree, T.I. Phenotypic complexity of the human regulatory T cell compartment revealed by mass cytometry. J. Immunol. 2015, 195, 2030–2037. [Google Scholar] [CrossRef] [PubMed]
  108. Yamaguchi, T.; Wing, J.B.; Sakaguchi, S. Two modes of immune suppression by Foxp3+ regulatory T cells under inflammatory or non-inflammatory conditions. Semin. Immunol. 2011, 23, 424–430. [Google Scholar] [CrossRef]
  109. Vignali, D.A. Mechanisms of Treg suppression: Still a long way to go. Front. Immunol. 2012, 3, 191. [Google Scholar] [CrossRef] [PubMed]
  110. Markov, A.G.; Aschenbach, J.R.; Amasheh, S. The epithelial barrier and beyond: Claudins as amplifiers of physiological organ functions. IUBMB Life 2017, 69, 290–296. [Google Scholar] [CrossRef]
  111. Snoeck, V.; Goddeeris, B.; Cox, E. The role of enterocytes in the intestinal barrier function and antigen uptake. Microbes Infect. 2005, 7, 997–1004. [Google Scholar] [CrossRef] [PubMed]
  112. Lechuga, S.; Braga-Neto, M.B.; Naydenov, N.G.; Rieder, F.; Ivanov, A.I. Understanding disruption of the gut barrier during inflammation: Should we abandon traditional epithelial cell lines and switch to intestinal organoids? Front. Immunol. 2023, 14, 1108289. [Google Scholar] [CrossRef] [PubMed]
  113. Buckley, A.; Turner, J.R. Cell biology of tight junction barrier regulation and mucosal disease. Cold Spring Harb. Perspect. Biol. 2018, 10, a029314. [Google Scholar] [CrossRef] [PubMed]
  114. Laukoetter, M.G.; Bruewer, M.; Nusrat, A. Regulation of the intestinal epithelial barrier by the apical junctional complex. Curr. Opin. Gastroenterol. 2006, 22, 85–89. [Google Scholar] [CrossRef]
  115. Guo, C.; Shen, J. Cytoskeletal organization and cell polarity in the pathogenesis of Crohn’s disease. Clin. Rev. Allergy Immunol. 2021, 60, 164–174. [Google Scholar] [CrossRef]
  116. Lechuga, S.; Ivanov, A.I. Disruption of the epithelial barrier during intestinal inflammation: Quest for new molecules and mechanisms. Biochim. Et Biophys. Acta Mol. Cell Res. 2017, 1864, 1183–1194. [Google Scholar] [CrossRef]
  117. Ivanov, A.I.; Parkos, C.A.; Nusrat, A. Cytoskeletal regulation of epithelial barrier function during inflammation. Am. J. Pathol. 2010, 177, 512–524. [Google Scholar] [CrossRef]
  118. Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.-D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability–a new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef]
  119. Lopetuso, L.R.; Scaldaferri, F.; Bruno, G.; Petito, V.; Franceschi, F.; Gasbarrini, A. The therapeutic management of gut barrier leaking: The emerging role for mucosal barrier protectors. Eur. Rev. Med. Pharmacol. Sci. 2015, 9, 1068–1076. [Google Scholar]
  120. Iweala, O.I.; Nagler, C.R. The microbiome and food allergy. Annu. Rev. Immunol. 2019, 37, 377–403. [Google Scholar] [CrossRef] [PubMed]
  121. Odenwald, M.A.; Turner, J.R. The intestinal epithelial barrier: A therapeutic target? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 9–21. [Google Scholar] [CrossRef]
  122. Zimmermann, P.; Messina, N.; Mohn, W.W.; Finlay, B.B.; Curtis, N. Association between the intestinal microbiota and allergic sensitization, eczema, and asthma: A systematic review. J. Allergy Clin. Immunol. 2019, 143, 467–485. [Google Scholar] [CrossRef]
  123. Liang, L.; Saunders, C.; Sanossian, N. Food, gut barrier dysfunction, and related diseases: A new target for future individualized disease prevention and management. Food Sci. Nutr. 2023, 11, 1671–1704. [Google Scholar] [CrossRef]
  124. Maeda, K.; Caldez, M.J.; Akira, S. Innate immunity in allergy. Allergy 2019, 74, 1660–1674. [Google Scholar] [CrossRef]
  125. Loh, W.; Tang, M.L. The epidemiology of food allergy in the global context. Int. J. Environ. Res. Public Health 2018, 15, 2043. [Google Scholar] [CrossRef] [PubMed]
  126. Berni Canani, R.; Paparo, L.; Nocerino, R.; Di Scala, C.; Della Gatta, G.; Maddalena, Y.; Buono, A.; Bruno, C.; Voto, L.; Ercolini, D. Gut microbiome as target for innovative strategies against food allergy. Front. Immunol. 2019, 10, 191. [Google Scholar] [CrossRef]
  127. De Filippis, F.; Paparo, L.; Nocerino, R.; Della Gatta, G.; Carucci, L.; Russo, R.; Pasolli, E.; Ercolini, D.; Berni Canani, R. Specific gut microbiome signatures and the associated pro-inflammatory functions are linked to pediatric allergy and acquisition of immune tolerance. Nat. Commun. 2021, 12, 5958. [Google Scholar] [CrossRef]
  128. Wilkins, L.J.; Monga, M.; Miller, A.W. Defining dysbiosis for a cluster of chronic diseases. Sci. Rep. 2019, 9, 12918. [Google Scholar] [CrossRef]
  129. Lee, K.H.; Song, Y.; Wu, W.; Yu, K.; Zhang, G. The gut microbiota, environmental factors, and links to the development of food allergy. Clin. Mol. Allergy 2020, 18, 5. [Google Scholar] [CrossRef] [PubMed]
  130. Cait, A.; Cardenas, E.; Dimitriu, P.A.; Amenyogbe, N.; Dai, D.; Cait, J.; Sbihi, H.; Stiemsma, L.; Subbarao, P.; Mandhane, P.J. Reduced genetic potential for butyrate fermentation in the gut microbiome of infants who develop allergic sensitization. J. Allergy Clin. Immunol. 2019, 144, 1638–1647.e3. [Google Scholar] [CrossRef]
  131. Feehley, T.; Plunkett, C.H.; Bao, R.; Choi Hong, S.M.; Culleen, E.; Belda-Ferre, P.; Campbell, E.; Aitoro, R.; Nocerino, R.; Paparo, L. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat. Med. 2019, 25, 448–453. [Google Scholar] [CrossRef]
  132. Berni Canani, R.; Sangwan, N.; Stefka, A.T.; Nocerino, R.; Paparo, L.; Aitoro, R.; Calignano, A.; Khan, A.A.; Gilbert, J.A.; Nagler, C.R. Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. ISME J. 2016, 10, 742–750. [Google Scholar] [CrossRef] [PubMed]
  133. Chua, H.-H.; Chou, H.-C.; Tung, Y.-L.; Chiang, B.-L.; Liao, C.-C.; Liu, H.-H.; Ni, Y.-H. Intestinal dysbiosis featuring abundance of Ruminococcus gnavus associates with allergic diseases in infants. Gastroenterology 2018, 154, 154–167. [Google Scholar] [CrossRef]
  134. Galazzo, G.; van Best, N.; Bervoets, L.; Dapaah, I.O.; Savelkoul, P.H.; Hornef, M.W.; Hutton, E.K.; Morrison, K.; Holloway, A.C.; McDonald, H. Development of the microbiota and associations with birth mode, diet, and atopic disorders in a longitudinal analysis of stool samples, collected from infancy through early childhood. Gastroenterology 2020, 158, 1584–1596. [Google Scholar] [CrossRef] [PubMed]
  135. Bao, R.; Hesser, L.A.; He, Z.; Zhou, X.; Nadeau, K.C.; Nagler, C.R. Fecal microbiome and metabolome differ in healthy and food-allergic twins. J. Clin. Investig. 2021, 131, e141935. [Google Scholar] [CrossRef] [PubMed]
  136. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-y, M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef]
  137. Paparo, L.; Nocerino, R.; Ciaglia, E.; Di Scala, C.; De Caro, C.; Russo, R.; Trinchese, G.; Aitoro, R.; Amoroso, A.; Bruno, C. Butyrate as a bioactive human milk protective component against food allergy. Allergy 2021, 76, 1398–1415. [Google Scholar] [CrossRef]
  138. Depner, M.; Taft, D.H.; Kirjavainen, P.V.; Kalanetra, K.M.; Karvonen, A.M.; Peschel, S.; Schmausser-Hechfellner, E.; Roduit, C.; Frei, R.; Lauener, R. Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nat. Med. 2020, 26, 1766–1775. [Google Scholar] [CrossRef]
  139. Goldberg, M.R.; Mor, H.; Magid Neriya, D.; Magzal, F.; Muller, E.; Appel, M.Y.; Nachshon, L.; Borenstein, E.; Tamir, S.; Louzoun, Y. Microbial signature in IgE-mediated food allergies. Genome Med. 2020, 12, 92. [Google Scholar] [CrossRef] [PubMed]
  140. Berni Canani, R.; De Filippis, F.; Nocerino, R.; Paparo, L.; Di Scala, C.; Cosenza, L.; Della Gatta, G.; Calignano, A.; De Caro, C.; Laiola, M. Gut microbiota composition and butyrate production in children affected by non-IgE-mediated cow’s milk allergy. Sci. Rep. 2018, 8, 12500. [Google Scholar] [CrossRef]
  141. Shim, J.A.; Ryu, J.H.; Jo, Y.; Hong, C. The role of gut microbiota in T cell immunity and immune mediated disorders. Int. J. Biol. Sci. 2023, 19, 1178. [Google Scholar] [CrossRef]
  142. Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 2010, 107, 12204–12209. [Google Scholar] [CrossRef]
  143. Stefka, A.T.; Feehley, T.; Tripathi, P.; Qiu, J.; McCoy, K.; Mazmanian, S.K.; Tjota, M.Y.; Seo, G.-Y.; Cao, S.; Theriault, B.R. Commensal bacteria protect against food allergen sensitization. Proc. Natl. Acad. Sci. USA 2014, 111, 13145–13150. [Google Scholar] [CrossRef] [PubMed]
  144. Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
  145. Savage, J.H.; Lee-Sarwar, K.A.; Sordillo, J.; Bunyavanich, S.; Zhou, Y.; O’Connor, G.; Sandel, M.; Bacharier, L.B.; Zeiger, R.; Sodergren, E.; et al. A prospective microbiome-wide association study of food sensitization and food allergy in early childhood. Allergy 2018, 73, 145–152. [Google Scholar] [CrossRef] [PubMed]
  146. Cheng, Y.; Liu, X.; Chen, F.; Rolnik, B.M.; Chleilat, F.; Ling, Z.; Snyder, M.P.; Zhou, X. The Roles and Mechanisms of Gut Microbiota in Food Allergy. Adv. Gut Microbiome Res. 2023, 2023, 9575410. [Google Scholar] [CrossRef]
  147. Yang, H.; Qu, Y.; Gao, Y.; Sun, S.; Wu, R.; Wu, J. Research Progress on the Correlation between the Intestinal Microbiota and Food Allergy. Foods 2022, 11, 2913. [Google Scholar] [CrossRef]
  148. Madhogaria, B.; Bhowmik, P.; Kundu, A. Correlation between human gut microbiome and diseases. Infect. Med. 2022, 1, 180–191. [Google Scholar] [CrossRef]
  149. Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [PubMed]
  150. Bunyavanich, S.; Shen, N.; Grishin, A.; Wood, R.; Burks, W.; Dawson, P.; Jones, S.M.; Leung, D.Y.M.; Sampson, H.; Sicherer, S.; et al. Early-life gut microbiome composition and milk allergy resolution. J. Allergy Clin. Immunol. 2016, 138, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  151. Thompson-Chagoyan, O.C.; Vieites, J.M.; Maldonado, J.; Edwards, C.; Gil, A. Changes in faecal microbiota of infants with cow’s milk protein allergy—A Spanish prospective case-control 6-month follow-up study. Pediatr. Allergy Immunol. 2010, 21, e394–e400. [Google Scholar] [CrossRef]
  152. Fazlollahi, M.; Chun, Y.; Grishin, A.; Wood, R.A.; Burks, A.W.; Dawson, P.; Jones, S.M.; Leung, D.Y.M.; Sampson, H.A.; Sicherer, S.H.; et al. Early-life gut microbiome and egg allergy. Allergy 2018, 73, 1515–1524. [Google Scholar] [CrossRef] [PubMed]
  153. Fric, P.; Gabrovska, D.; Nevoral, J. Celiac disease, gluten-free diet, and oats. Nutr. Rev. 2011, 69, 107–115. [Google Scholar] [CrossRef]
  154. Aljada, B.; Zohni, A.; El-Matary, W. The Gluten-Free Diet for Celiac Disease and Beyond. Nutrients 2021, 13, 3993. [Google Scholar] [CrossRef] [PubMed]
  155. De Palma, G.; Nadal, I.; Collado, M.C.; Sanz, Y. Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects. Br. J. Nutr. 2009, 102, 1154–1160. [Google Scholar] [CrossRef]
  156. Hemarajata, P.; Versalovic, J. Effects of probiotics on gut microbiota: Mechanisms of intestinal immunomodulation and neuromodulation. Ther. Adv. Gastroenterol. 2013, 6, 39–51. [Google Scholar] [CrossRef]
  157. Yang, B.; Xiao, L.; Liu, S.; Liu, X.; Luo, Y.; Ji, Q.; Yang, P.; Liu, Z. Exploration of the effect of probiotics supplementation on intestinal microbiota of food allergic mice. Am. J. Transl. Res. 2017, 9, 376. [Google Scholar]
  158. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 61, 1600240. [Google Scholar] [CrossRef]
  159. Caminero, A.; Meisel, M.; Jabri, B.; Verdu, E.F. Mechanisms by which gut microorganisms influence food sensitivities. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 7–18. [Google Scholar] [CrossRef] [PubMed]
  160. Caminero, A.; Herrán, A.R.; Nistal, E.; Pérez-Andrés, J.; Vaquero, L.; Vivas, S.; Ruiz de Morales, J.M.G.; Albillos, S.M.; Casqueiro, J. Diversity of the cultivable human gut microbiome involved in gluten metabolism: Isolation of microorganisms with potential interest for coeliac disease. FEMS Microbiol. Ecol. 2014, 88, 309–319. [Google Scholar] [CrossRef] [PubMed]
  161. Caminero, A.; Nistal, E.; Herrán, A.R.; Pérez-Andrés, J.; Ferrero, M.A.; Ayala, L.V.; Vivas, S.; de Morales, J.M.R.; Albillos, S.M.; Casqueiro, F.J. Differences in gluten metabolism among healthy volunteers, coeliac disease patients and first-degree relatives. Br. J. Nutr. 2015, 114, 1157–1167. [Google Scholar] [CrossRef]
  162. Herrán, A.R.; Pérez-Andrés, J.; Caminero, A.; Nistal, E.; Vivas, S.; de Morales, J.M.R.; Casqueiro, J. Gluten-degrading bacteria are present in the human small intestine of healthy volunteers and celiac patients. Res. Microbiol. 2017, 168, 673–684. [Google Scholar] [CrossRef] [PubMed]
  163. Helmerhorst, E.J.; Zamakhchari, M.; Schuppan, D.; Oppenheim, F.G. Discovery of a novel and rich source of gluten-degrading microbial enzymes in the oral cavity. PLoS ONE 2010, 5, e13264. [Google Scholar] [CrossRef]
  164. Fernandez-Feo, M.; Wei, G.; Blumenkranz, G.; Dewhirst, F.E.; Schuppan, D.; Oppenheim, F.G.; Helmerhorst, E.J. The cultivable human oral gluten-degrading microbiome and its potential implications in coeliac disease and gluten sensitivity. Clin. Microbiol. Infect. 2013, 19, E386–E394. [Google Scholar] [CrossRef]
  165. Dowling, D.J.; Levy, O. Ontogeny of early life immunity. Trends Immunol. 2014, 35, 299–310. [Google Scholar] [CrossRef]
  166. Julia, V.; Macia, L.; Dombrowicz, D. The impact of diet on asthma and allergic diseases. Nat. Rev. Immunol. 2015, 15, 308–322. [Google Scholar] [CrossRef]
  167. Wang, S.; Yin, P.; Yu, L.; Tian, F.; Chen, W.; Zhai, Q. Effects of Early Diet on the Prevalence of Allergic Disease in Children: A Systematic Review and Meta-Analysis. Adv. Nutr. 2024, 15, 100128. [Google Scholar] [CrossRef]
  168. Sansotta, N.; Piacentini, G.L.; Mazzei, F.; Minniti, F.; Boner, A.L.; Peroni, D.G. Timing of introduction of solid food and risk of allergic disease development: Understanding the evidence. Allergol. Immunopathol. 2013, 41, 337–345. [Google Scholar] [CrossRef]
  169. Scarpone, R.; Kimkool, P.; Ierodiakonou, D.; Leonardi-Bee, J.; Garcia-Larsen, V.; Perkin, M.R.; Boyle, R.J. Timing of allergenic food introduction and risk of immunoglobulin E–mediated food allergy: A systematic review and meta-analysis. JAMA Pediatr. 2023, 177, 489–497. [Google Scholar] [CrossRef]
  170. Skjerven, H.O.; Lie, A.; Vettukattil, R.; Rehbinder, E.M.; LeBlanc, M.; Asarnoj, A.; Carlsen, K.-H.; Despriee, Å.W.; Färdig, M.; Gerdin, S.W. Early food intervention and skin emollients to prevent food allergy in young children (PreventADALL): A factorial, multicentre, cluster-randomised trial. Lancet 2022, 399, 2398–2411. [Google Scholar] [CrossRef] [PubMed]
  171. Halken, S.; Muraro, A.; de Silva, D.; Khaleva, E.; Angier, E.; Arasi, S.; Arshad, H.; Bahnson, H.T.; Beyer, K.; Boyle, R. EAACI guideline: Preventing the development of food allergy in infants and young children (2020 update). Pediatr. Allergy Immunol. 2021, 32, 843–858. [Google Scholar] [CrossRef]
  172. Perkin, M.R.; Logan, K.; Tseng, A.; Raji, B.; Ayis, S.; Peacock, J.; Brough, H.; Marrs, T.; Radulovic, S.; Craven, J. Randomized trial of introduction of allergenic foods in breast-fed infants. N. Engl. J. Med. 2016, 374, 1733–1743. [Google Scholar] [CrossRef] [PubMed]
  173. Perkin, M.R.; Logan, K.; Marrs, T.; Radulovic, S.; Craven, J.; Flohr, C.; Lack, G.; Young, L.; Offord, V.; DeSousa, M. Enquiring About Tolerance (EAT) study: Feasibility of an early allergenic food introduction regimen. J. Allergy Clin. Immunol. 2016, 137, 1477–1486.e8. [Google Scholar] [CrossRef] [PubMed]
  174. Adam, T.; Divaret-Chauveau, A.; Roduit, C.; Adel-Patient, K.; Deschildre, A.; Raherison, C.; Charles, M.A.; Nicklaus, S.; de Lauzon-Guillain, B. Complementary feeding practices are related to the risk of food allergy in the ELFE cohort. Allergy 2023, 78, 2456–2466. [Google Scholar] [CrossRef] [PubMed]
  175. Cleveland Clinic. Food Allergy vs. Intolerance: What’s the Difference? Available online: https://health.clevelandclinic.org/allergy-or-intolerance-how-to-tell-the-difference (accessed on 27 July 2024).
  176. Boyce, J.A.; Assa’ad, A.; Burks, A.W.; Jones, S.M.; Sampson, H.A.; Wood, R.A.; Plaut, M.; Cooper, S.F.; Fenton, M.J.; Arshad, S.H.; et al. Guidelines for the diagnosis and management of food allergy in the United States: Report of the NIAID-sponsored expert panel. J. Allergy Clin. Immunol. 2010, 126, S1–S58. [Google Scholar] [CrossRef]
  177. AllergyUK. Allergy vs. Intolerance. Available online: https://www.allergyuk.org/about-allergy/allergy-vs-intolerance/ (accessed on 27 July 2024).
  178. Zopf, Y.; Hahn, E.G.; Raithel, M.; Baenkler, H.-W.; Silbermann, A. The differential diagnosis of food intolerance. Dtsch. Ärzteblatt Int. 2009, 106, 359. [Google Scholar] [CrossRef]
  179. Young, E.; Stoneham, M.D.; Petruckevitch, A.; Barton, J.; Rona, R. A population study of food intolerance. Lancet 1994, 343, 1127–1130. [Google Scholar] [CrossRef]
  180. Bohn, L.; Storsrud, S.; Tornblom, H.; Bengtsson, U.J.; Simren, M. Tu2079 Food-Related Gastrointestinal Symptoms in IBS Are Common and Associated With More Severe Symptoms and Reduced Quality of Life. Gastroenterology 2013, 108, 634–641. [Google Scholar] [CrossRef]
  181. Hayes, P.A.; Fraher, M.H.; Quigley, E.M. Irritable bowel syndrome: The role of food in pathogenesis and management. Gastroenterol. Hepatol. 2014, 10, 164. [Google Scholar]
  182. Monsbakken, K.W.; Vandvik, P.O.; Farup, P.G. Perceived food intolerance in subjects with irritable bowel syndrome–etiology, prevalence and consequences. Eur. J. Clin. Nutr. 2006, 60, 667–672. [Google Scholar] [CrossRef]
  183. Boyce, J.A.; Assa’ad, A.; Burks, A.W.; Jones, S.M.; Sampson, H.A.; Wood, R.A.; Plaut, M.; Cooper, S.F.; Fenton, M.J.; Arshad, S.H.; et al. Guidelines for the diagnosis and management of food allergy in the United States: Summary of the NIAID-sponsored expert panel report. Nutr. Res. 2011, 31, 61–75. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  184. Gargano, D.; Appanna, R.; Santonicola, A.; De Bartolomeis, F.; Stellato, C.; Cianferoni, A.; Casolaro, V.; Iovino, P. Food Allergy and Intolerance: A Narrative Review on Nutritional Concerns. Nutrients 2021, 13, 1638. [Google Scholar] [CrossRef] [PubMed]
  185. Caminero, A.; Galipeau, H.J.; McCarville, J.L.; Johnston, C.W.; Bernier, S.P.; Russell, A.K.; Jury, J.; Herran, A.R.; Casqueiro, J.; Tye-Din, J.A. Duodenal bacteria from patients with celiac disease and healthy subjects distinctly affect gluten breakdown and immunogenicity. Gastroenterology 2016, 151, 670–683. [Google Scholar] [CrossRef]
  186. Galipeau, H.J.; McCarville, J.L.; Huebener, S.; Litwin, O.; Meisel, M.; Jabri, B.; Sanz, Y.; Murray, J.A.; Jordana, M.; Alaedini, A. Intestinal microbiota modulates gluten-induced immunopathology in humanized mice. Am. J. Pathol. 2015, 185, 2969–2982. [Google Scholar] [CrossRef] [PubMed]
  187. Halmos, E.P.; Power, V.A.; Shepherd, S.J.; Gibson, P.R.; Muir, J.G. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology 2014, 146, 67–75.e5. [Google Scholar] [CrossRef] [PubMed]
  188. Tuck, C.J.; Biesiekierski, J.R.; Schmid-Grendelmeier, P.; Pohl, D. Food Intolerances. Nutrients 2019, 11, 1684. [Google Scholar] [CrossRef] [PubMed]
  189. Lomer, M.C.; Parkes, G.; Sanderson, J. Lactose intolerance in clinical practice–myths and realities. Aliment. Pharmacol. Ther. 2008, 27, 93–103. [Google Scholar] [CrossRef]
  190. Bonder, M.J.; Kurilshikov, A.; Tigchelaar, E.F.; Mujagic, Z.; Imhann, F.; Vila, A.V.; Deelen, P.; Vatanen, T.; Schirmer, M.; Smeekens, S.P. The effect of host genetics on the gut microbiome. Nat. Genet. 2016, 48, 1407–1412. [Google Scholar] [CrossRef]
  191. Brandao Gois, M.F.; Sinha, T.; Spreckels, J.E.; Vich Vila, A.; Bolte, L.A.; Weersma, R.K.; Wijmenga, C.; Fu, J.; Zhernakova, A.; Kurilshikov, A. Role of the gut microbiome in mediating lactose intolerance symptoms. Gut 2022, 71, 215–217. [Google Scholar] [CrossRef]
  192. He, T.; Venema, K.; Priebe, M.; Welling, G.; Brummer, R.J.; Vonk, R. The role of colonic metabolism in lactose intolerance. Eur. J. Clin. Investig. 2008, 38, 541–547. [Google Scholar] [CrossRef]
  193. He, T.; Priebe, M.G.; Harmsen, H.J.; Stellaard, F.; Sun, X.; Welling, G.W.; Vonk, R.J. Colonic fermentation may play a role in lactose intolerance in humans. J. Nutr. 2006, 136, 58–63. [Google Scholar] [CrossRef] [PubMed]
  194. Staudacher, H.M.; Lomer, M.C.; Farquharson, F.M.; Louis, P.; Fava, F.; Franciosi, E.; Scholz, M.; Tuohy, K.M.; Lindsay, J.O.; Irving, P.M. A diet low in FODMAPs reduces symptoms in patients with irritable bowel syndrome and a probiotic restores Bifidobacterium species: A randomized controlled trial. Gastroenterology 2017, 153, 936–947. [Google Scholar] [CrossRef]
  195. Mysore Saiprasad, S.; Moreno, O.G.; Savaiano, D.A. A Narrative Review of Human Clinical Trials to Improve Lactose Digestion and Tolerance by Feeding Bifidobacteria or Galacto-Oligosacharides. Nutrients 2023, 15, 3559. [Google Scholar] [CrossRef] [PubMed]
  196. Wielgosz-Grochowska, J.P.; Domanski, N.; Drywien, M.E. Identification of SIBO Subtypes along with Nutritional Status and Diet as Key Elements of SIBO Therapy. Int. J. Mol. Sci. 2024, 25, 7341. [Google Scholar] [CrossRef]
  197. Knez, E.; Kadac-Czapska, K.; Grembecka, M. The importance of food quality, gut motility, and microbiome in SIBO development and treatment. Nutrition 2024, 124, 112464. [Google Scholar] [CrossRef] [PubMed]
  198. Lu, S.; Chen, Y.; Guo, H.; Liu, Z.; Du, Y.; Duan, L. Differences in clinical manifestations and the fecal microbiome between irritable bowel syndrome and small intestinal bacterial overgrowth. Dig. Liver Dis. 2024. [Google Scholar] [CrossRef] [PubMed]
  199. Ames, S.R.; Lotoski, L.C.; Azad, M.B. Comparing early life nutritional sources and human milk feeding practices: Personalized and dynamic nutrition supports infant gut microbiome development and immune system maturation. Gut Microbes 2023, 15, 2190305. [Google Scholar] [CrossRef]
  200. Widstrom, A.M.; Brimdyr, K.; Svensson, K.; Cadwell, K.; Nissen, E. Skin-to-skin contact the first hour after birth, underlying implications and clinical practice. Acta Paediatr. 2019, 108, 1192–1204. [Google Scholar] [CrossRef]
  201. Davanzo, R.; Strajn, T.; Kennedy, J.; Crocetta, A.; De Cunto, A. From tube to breast: The bridging role of semi-demand breastfeeding. J. Hum. Lact. 2014, 30, 405–409. [Google Scholar] [CrossRef] [PubMed]
  202. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiol. Open 2022, 11, e1260. [Google Scholar] [CrossRef]
  203. Bzikowska-Jura, A.; Czerwonogrodzka-Senczyna, A.; Oledzka, G.; Szostak-Wegierek, D.; Weker, H.; Wesolowska, A. Maternal Nutrition and Body Composition During Breastfeeding: Association with Human Milk Composition. Nutrients 2018, 10, 1379. [Google Scholar] [CrossRef] [PubMed]
  204. Savage, J.H.; Lee-Sarwar, K.A.; Sordillo, J.E.; Lange, N.E.; Zhou, Y.; O’Connor, G.T.; Sandel, M.; Bacharier, L.B.; Zeiger, R.; Sodergren, E.; et al. Diet during Pregnancy and Infancy and the Infant Intestinal Microbiome. J. Pediatr. 2018, 203, 47–54.e4. [Google Scholar] [CrossRef]
  205. van den Akker, C.H.P.; van Goudoever, J.B.; Shamir, R.; Domellof, M.; Embleton, N.D.; Hojsak, I.; Lapillonne, A.; Mihatsch, W.A.; Berni Canani, R.; Bronsky, J.; et al. Probiotics and Preterm Infants: A Position Paper by the European Society for Paediatric Gastroenterology Hepatology and Nutrition Committee on Nutrition and the European Society for Paediatric Gastroenterology Hepatology and Nutrition Working Group for Probiotics and Prebiotics. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 664–680. [Google Scholar] [CrossRef]
  206. Ruiz-Ojeda, F.J.; Plaza-Diaz, J.; Morales, J.; Alvarez-Calatayud, G.; Climent, E.; Silva, A.; Martinez-Blanch, J.F.; Enrique, M.; Tortajada, M.; Ramon, D.; et al. Effects of a Novel Infant Formula on the Fecal Microbiota in the First Six Months of Life: The INNOVA 2020 Study. Int. J. Mol. Sci. 2023, 24, 3034. [Google Scholar] [CrossRef] [PubMed]
  207. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Morales, J.; Martin-Masot, R.; Climent, E.; Silva, A.; Martinez-Blanch, J.F.; Enrique, M.; Tortajada, M.; Ramon, D.; et al. Innova 2020: A Follow-Up Study of the Fecal Microbiota of Infants Using a Novel Infant Formula between 6 Months and 12 Months of Age. Int. J. Mol. Sci. 2023, 24, 7392. [Google Scholar] [CrossRef] [PubMed]
  208. Martinez Suarez, V. Use of probiotics and prebiotics in infant formulas. Nutr. Hosp. 2015, 31 (Suppl. S1), 72–77. [Google Scholar] [CrossRef] [PubMed]
  209. Adamczyk, I.; Kaliszczak, K.; Skowron, K.; Grudlewska-Buda, K.; Twaruzek, M.; Sinkiewicz-Darol, E. Microbiological status of donor human milk—A single center study from Poland. Food Microbiol. 2024, 122, 104528. [Google Scholar] [CrossRef]
  210. Aktas Reyhan, F. The effect of breastfeeding education with digital storytelling on fathers’ breastfeeding self-efficacy. J. Eval. Clin. Pract. 2024. early view. [Google Scholar] [CrossRef]
  211. Bai, R.; Cheng, Y.; Shan, S.; Zhao, X.; Wei, J.; Xia, C. The breastfeeding experience of women with multiple pregnancies: A meta-synthesis of qualitative studies. BMC Pregnancy Childbirth 2024, 24, 492. [Google Scholar] [CrossRef] [PubMed]
  212. Hiito, E.; Ikonen, R.; Niela-Vilen, H. Internet-based breastfeeding peer support for breastfeeding parents: An integrative review. J. Adv. Nurs. 2024. early view. [Google Scholar] [CrossRef]
  213. Syahri, I.M.; Laksono, A.D.; Fitria, M.; Rohmah, N.; Masruroh, M.; Ipa, M. Exclusive breastfeeding among Indonesian working mothers: Does early initiation of breastfeeding matter? BMC Public Health 2024, 24, 1225. [Google Scholar] [CrossRef] [PubMed]
  214. Fair, F.J.; Morison, A.; Soltani, H. Stakeholders’ views of the Baby Friendly Initiative implementation and impact: A mixed methods study. Int. Breastfeed. J. 2024, 19, 49. [Google Scholar] [CrossRef] [PubMed]
  215. Souza, C.B.; Melo, D.S.; Relvas, G.R.B.; Venancio, S.I.; Silva, R. Promotion, protection, and support of breastfeeding at work, and achieving sustainable development: A scoping review. Cien Saude Colet. 2023, 28, 1059–1072. [Google Scholar] [CrossRef] [PubMed]
  216. Hill, Z.; Manu, A.; Tawiah-Agyemang, C.; Gyan, T.; Turner, K.; Weobong, B.; Ten Asbroek, A.H.; Kirkwood, B.R. How did formative research inform the development of a home-based neonatal care intervention in rural Ghana? J. Perinatol. 2008, 28 (Suppl. S2), S38–S45. [Google Scholar] [CrossRef]
  217. Dann, M.H. The lactation consult: Problem solving, teaching, and support for the breastfeeding family. J. Pediatr. Health Care 2005, 19, 12–16. [Google Scholar] [CrossRef]
  218. Masum, A.K.M.; Chandrapala, J.; Huppertz, T.; Adhikari, B.; Zisu, B. Production and characterization of infant milk formula powders: A review. Dry. Technol. 2021, 39, 1492–1512. [Google Scholar] [CrossRef]
  219. Hageman, J.H.J.; Danielsen, M.; Nieuwenhuizen, A.G.; Feitsma, A.L.; Dalsgaard, T.K. Comparison of bovine milk fat and vegetable fat for infant formula: Implications for infant health. Int. Dairy J. 2019, 92, 37–49. [Google Scholar] [CrossRef]
  220. Lund, P.; Bechshoft, M.R.; Ray, C.A.; Lund, M.N. Effect of Processing of Whey Protein Ingredient on Maillard Reactions and Protein Structural Changes in Powdered Infant Formula. J. Agric. Food Chem. 2022, 70, 319–332. [Google Scholar] [CrossRef]
  221. Imdad, A.; Sherwani, R.; Wall, K. Pediatric Formulas: An Update. Pediatr. Rev. 2024, 45, 394–405. [Google Scholar] [CrossRef]
  222. Prosser, C.G. Compositional and functional characteristics of goat milk and relevance as a base for infant formula. J. Food Sci. 2021, 86, 257–265. [Google Scholar] [CrossRef] [PubMed]
  223. Migacheva, N.B.; Mukhametova, E.M.; Makarova, E.G.; Ukraintsev, S.E. The Role and Place of Partially Hydrolysed Protein Infant Formulas in the Nutrition of Full-term Children: Digestive Comfort and Allergy Prevention. Curr. Pediatr. 2020, 19, 279–290. [Google Scholar] [CrossRef]
  224. Juliano, B.O. Rice: Chemistry and Technology; Woodhead Publishing: Sawston, UK, 1985. [Google Scholar]
  225. Dupont, C.; Bocquet, A.; Tome, D.; Bernard, M.; Campeotto, F.; Dumond, P.; Essex, A.; Frelut, M.L.; Guenard-Bilbault, L.; Lack, G.; et al. Hydrolyzed Rice Protein-Based Formulas, a Vegetal Alternative in Cow’s Milk Allergy. Nutrients 2020, 12, 2654. [Google Scholar] [CrossRef]
  226. Bocquet, A.; Dupont, C.; Chouraqui, J.-P.; Darmaun, D.; Feillet, F.; Frelut, M.-L.; Girardet, J.-P.; Hankard, R.; Lapillonne, A.; Rozé, J.-C. Efficacy and safety of hydrolyzed rice-protein formulas for the treatment of cow’s milk protein allergy. Arch. Pédiatrie 2019, 26, 238–246. [Google Scholar] [CrossRef] [PubMed]
  227. Fiocchi, A.; Barrio-Torres, J.; Dupont, C.; Howells, H.E.; Shamir, R.; Venter, C.; Meyer, R. Hydrolyzed rice formula for dietary management of infants with cow’s milk allergy. World Allergy Organ. J. 2022, 15, 100717. [Google Scholar] [CrossRef]
  228. Maryniak, N.Z.; Sancho, A.I.; Hansen, E.B.; Bogh, K.L. Alternatives to Cow’s Milk-Based Infant Formulas in the Prevention and Management of Cow’s Milk Allergy. Foods 2022, 11, 926. [Google Scholar] [CrossRef] [PubMed]
  229. Milbrandt, T.P. Specialized Infant Formulas. Pediatr. Rev. 2017, 38, 241–242. [Google Scholar] [CrossRef]
  230. Martinez, J.A.; Ballew, M.P. Infant formulas. Pediatr. Rev. 2011, 32, 179–189, quiz 189. [Google Scholar] [CrossRef]
  231. Plaza-Diaz, J.; Fontana, L.; Gil, A. Human Milk Oligosaccharides and Immune System Development. Nutrients 2018, 10, 1038. [Google Scholar] [CrossRef]
  232. Kunz, C. Historical aspects of human milk oligosaccharides. Adv. Nutr. 2012, 3, 430S–439S. [Google Scholar] [CrossRef] [PubMed]
  233. Bode, L. Human milk oligosaccharides: Every baby needs a sugar mama. Glycobiology 2012, 22, 1147–1162. [Google Scholar] [CrossRef]
  234. Jantscher-Krenn, E.; Bode, L. Human milk oligosaccharides and their potential benefits for the breast-fed neonate. Minerva Pediatr. 2012, 64, 83–99. [Google Scholar]
  235. Smilowitz, J.T.; Lebrilla, C.B.; Mills, D.A.; German, J.B.; Freeman, S.L. Breast milk oligosaccharides: Structure-function relationships in the neonate. Annu. Rev. Nutr. 2014, 34, 143–169. [Google Scholar] [CrossRef] [PubMed]
  236. 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]
  237. Dinleyici, M.; Barbieur, J.; Dinleyici, E.C.; Vandenplas, Y. Functional effects of human milk oligosaccharides (HMOs). Gut Microbes 2023, 15, 2186115. [Google Scholar] [CrossRef] [PubMed]
  238. Sprenger, N.; Tytgat, H.L.; Binia, A.; Austin, S.; Singhal, A. Biology of human milk oligosaccharides: From basic science to clinical evidence. J. Human Nutr. Diet. 2022, 35, 280–299. [Google Scholar] [CrossRef]
  239. Mosca, F.; Giannì, M.L. Human Milk: Composition and Health Benefits. Pediatr. Med. Chir. 2017, 39, 155. [Google Scholar] [CrossRef]
  240. Hill, D.R.; Chow, J.M.; Buck, R.H. Multifunctional Benefits of Prevalent HMOs: Implications for Infant Health. Nutrients 2021, 13, 3364. [Google Scholar] [CrossRef]
  241. European Society for Paediatric Gastroenterology; Hepatology & Nutrition (ESPGHAN); Fewtrell, M.; Baumann, U.; Bronsky, J.; Haiden, N.; Hill, S.; Kivelä, L.; de Koenig, B.; Köglmeier, J.; et al. World Health Organization (WHO) guideline on the complementary feeding of infants and young children aged 6-23 months 2023: A multisociety response. J. Pediatr. Gastroenterol. Nutr. 2024, 79, 181–188. [Google Scholar] [CrossRef]
  242. Lutter, C.K.; Grummer-Strawn, L.; Rogers, L. Complementary feeding of infants and young children 6 to 23 months of age. Nutr. Rev. 2021, 79, 825–846. [Google Scholar] [CrossRef] [PubMed]
  243. Clayton, H.B.; Li, R.; Perrine, C.G.; Scanlon, K.S. Prevalence and reasons for introducing infants early to solid foods: Variations by milk feeding type. Pediatrics 2013, 131, e1108–e1114. [Google Scholar] [CrossRef]
  244. 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] [PubMed]
  245. Coulthard, H.; Harris, G.; Emmett, P. Delayed introduction of lumpy foods to children during the complementary feeding period affects child’s food acceptance and feeding at 7 years of age. Matern. Child. Nutr. 2009, 5, 75–85. [Google Scholar] [CrossRef] [PubMed]
  246. de Paiva, E.L.; Ali, S.; Vasco, E.R.; Alvito, P.C.; de Oliveira, C.A.F. Bioaccessibility data of potentially toxic elements in complementary foods for infants: A review. Food Res. Int. 2023, 174, 113485. [Google Scholar] [CrossRef]
  247. Norris, J.M.; Barriga, K.; Hoffenberg, E.J.; Taki, I.; Miao, D.; Haas, J.E.; Emery, L.M.; Sokol, R.J.; Erlich, H.A.; Eisenbarth, G.S.; et al. Risk of celiac disease autoimmunity and timing of gluten introduction in the diet of infants at increased risk of disease. JAMA 2005, 293, 2343–2351. [Google Scholar] [CrossRef]
  248. Fangupo, L.J.; Heath, A.M.; Williams, S.M.; Erickson Williams, L.W.; Morison, B.J.; Fleming, E.A.; Taylor, B.J.; Wheeler, B.J.; Taylor, R.W. A Baby-Led Approach to Eating Solids and Risk of Choking. Pediatrics 2016, 138, e20160772. [Google Scholar] [CrossRef]
  249. Turnbull, J.L.; Adams, H.N.; Gorard, D.A. Review article: The diagnosis and management of food allergy and food intolerances. Aliment. Pharmacol. Ther. 2015, 41, 3–25. [Google Scholar] [CrossRef]
  250. Danielewicz, H. Breastfeeding and Allergy Effect Modified by Genetic, Environmental, Dietary, and Immunological Factors. Nutrients 2022, 14, 3011. [Google Scholar] [CrossRef]
  251. Wang, S.; Wei, Y.; Liu, L.; Li, Z. Association Between Breastmilk Microbiota and Food Allergy in Infants. Front. Cell Infect. Microbiol. 2021, 11, 770913. [Google Scholar] [CrossRef]
  252. Jadhav, A.; Bajaj, A.; Xiao, Y.; Markandey, M.; Ahuja, V.; Kashyap, P.C. Role of Diet-Microbiome Interaction in Gastrointestinal Disorders and Strategies to Modulate Them with Microbiome-Targeted Therapies. Annu. Rev. Nutr. 2023, 43, 355–383. [Google Scholar] [CrossRef]
  253. Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Verges, M.C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G.H.; et al. Microbiome definition re-visited: Old concepts and new challenges. Microbiome 2020, 8, 103. [Google Scholar] [CrossRef]
  254. Addae, H.Y.; Apprey, C.; Kwarteng, A. Gut Microbiome-Targeted Nutrition Interventions and Growth among Children in Low- and Middle-Income Countries: A Systematic Review and Meta-Analysis. Curr. Dev. Nutr. 2024, 8, 102085. [Google Scholar] [CrossRef] [PubMed]
  255. Zhang, X.; Li, L.; Butcher, J.; Stintzi, A.; Figeys, D. Advancing functional and translational microbiome research using meta-omics approaches. Microbiome 2019, 7, 154. [Google Scholar] [CrossRef]
  256. Mars, R.A.T.; Frith, M.; Kashyap, P.C. Functional Gastrointestinal Disorders and the Microbiome-What Is the Best Strategy for Moving Microbiome-based Therapies for Functional Gastrointestinal Disorders into the Clinic? Gastroenterology 2021, 160, 538–555. [Google Scholar] [CrossRef]
  257. Beller, L.; Matthijnssens, J. What is (not) known about the dynamics of the human gut virome in health and disease. Curr. Opin. Virol. 2019, 37, 52–57. [Google Scholar] [CrossRef] [PubMed]
  258. Wampach, L.; Heintz-Buschart, A.; Hogan, A.; Muller, E.E.L.; Narayanasamy, S.; Laczny, C.C.; Hugerth, L.W.; Bindl, L.; Bottu, J.; Andersson, A.F.; et al. Colonization and Succession within the Human Gut Microbiome by Archaea, Bacteria, and Microeukaryotes during the First Year of Life. Front. Microbiol. 2017, 8, 738. [Google Scholar] [CrossRef]
Figure 1. Changes in gut microbiome during infant feeding and in relation to allergies. Personal design.
Figure 1. Changes in gut microbiome during infant feeding and in relation to allergies. Personal design.
Children 11 01030 g001
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

Herrera-Quintana, L.; Vázquez-Lorente, H.; Hinojosa-Nogueira, D.; Plaza-Diaz, J. Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances. Children 2024, 11, 1030. https://doi.org/10.3390/children11081030

AMA Style

Herrera-Quintana L, Vázquez-Lorente H, Hinojosa-Nogueira D, Plaza-Diaz J. Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances. Children. 2024; 11(8):1030. https://doi.org/10.3390/children11081030

Chicago/Turabian Style

Herrera-Quintana, Lourdes, Héctor Vázquez-Lorente, Daniel Hinojosa-Nogueira, and Julio Plaza-Diaz. 2024. "Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances" Children 11, no. 8: 1030. https://doi.org/10.3390/children11081030

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