BMB Reports 2023; 56(9): 469-481  https://doi.org/10.5483/BMBRep.2023-0114
Scarring the early-life microbiome: its potential life-long effects on human health and diseases
Hyunji Park1,# , Na-Young Park1,# & Ara Koh1,2,*
1Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, 2Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
Correspondence to: Tel: +82-54-279-2320; Fax: +82-54-279-2199; E-mail: ara.koh@postech.ac.kr
#These authors contributed equally to this work.
Received: June 8, 2023; Revised: July 30, 2023; Accepted: August 14, 2023; Published online: August 30, 2023.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
The gut microbiome is widely recognized as a dynamic organ with a profound influence on human physiology and pathology. Extensive epidemiological and longitudinal cohort studies have provided compelling evidence that disruptions in the early-life microbiome can have long-lasting health implications. Various factors before, during, and after birth contribute to shaping the composition and function of the neonatal and infant microbiome. While these alterations can be partially restored over time, metabolic phenotypes may persist, necessitating research to identify the critical period for early intervention to achieve phenotypic recovery beyond microbiome composition. In this review, we provide current understanding of changes in the gut microbiota throughout life and the various factors affecting these changes. Specifically, we highlight the profound impact of early-life gut microbiota disruption on the development of diseases later in life and discuss perspectives on efforts to recover from such disruptions.
Keywords: Critical period, Early-life microbiome, Gut microbiome, Intervention, Life-long effects
INTRODUCTION

The human gut microbiome has a significant causal role in various aspects of human physiology and pathology, beyond its previously recognized associative roles. After birth, the human gut microbiome undergoes continuous changes, including increased microbial diversity, maturation and the emergence of site-specific microbial communities. While the composition of gut microbiota in adulthood is more stable than in infants, individual-specific gut microbiota shows personalized responses to diseases, dietary factors and drugs, indicating that individual microbiome may predict health outcomes (1). This raises a key question of when the impact of gut microbiome changes is the most significant.

Recent research has highlighted the significance of early-life gut microbiota in shaping host physiology, immunity and long-term health outcomes (2). The establishment of early gut microbiota is influenced by maternal factors during pregnancy, including microbiome transmission and metabolite transfer (3, 4). In addition, various factors such as gestational age, delivery mode and antibiotic exposure further impact the development and maturation of the early gut microbiota (5). Notably, the first 3 years from birth are considered a critical window for early intervention, as early-life colonization plays an important role in shaping long-term health outcomes even after the recovery from the gut microbial disruption (2, 6). Therefore, early intervention strategies are imperative to mitigate the long-lasting metabolic effects.

This review provides the basis for understanding the development of the gut microbiome in early life and emphasizes the importance of early interventions to normalize the microbiota from prenatal stages to 3 years after birth. These efforts aim not only to improve the health of infants but also to improve their long-term well-being.

GUT MICROBIOME IN HEALTH AND DISEASE

Altered gut microbiome in various human diseases

The adult human gut microbiota is a dynamic ecosystem that co-evolves with its host. It is estimated to consist of 39 trillion microbial cells, approximately the same number as human cells in our bodies (7). Moreover, this ecosystem has over a thousand times more genes than the human genome, indicating its significant genetic and metabolic potential and its substantial impact on the host (1). The gut microbiome has been linked to various human diseases, not limited to the gut, including inflammatory bowel disease and colon cancer, and neurological and metabolic diseases such as Parkinson’s disease, Alzheimer’s disease, autism, depression, type 2 diabetes, cardiovascular diseases, atherosclerosis, non-alcoholic fatty liver diseases and obesity (1, 8, 9). The gut microbiome’s influence on diseases beyond the gut is thought to occur through various microbial metabolites that enter the host’s bloodstream from the gut and reach different host tissues (9).

Dynamics of the normal gut microbiota

The interaction between the host and the gut microbial ecosystem is crucial for regulating the immune system and maintaining metabolic homeostasis, and altered gut microbial ecosystems are associated with various human diseases (10). However, alteration of the gut microbiota induced by many perturbations can be stochastic, and studies indicate that even under normal, ecologically stable conditions, fluctuations in the gut microbiome can occur (11). One of the challenges in investigating the association between gut microbiota and specific human diseases is the lack of consistent signatures. This inconsistency can be partly attributed to the limitations of cross-sectional cohort studies that fail to consider normal variations and proper control groups, as well as technical variations in sample collection and analysis (12). Therefore, longitudinal studies with repeated data collection could be important to identify consistent and specific microbial biomarkers by considering the temporal dynamics of the gut microbiota. In addition, age should be considered as a confounding factor in microbiome analysis results given that age-specific differences have been observed in gut microbial change-related diseases among individuals aged 20 to 89 years (13).

Recent longitudinal studies have shown that highly diverse gut microbial compositions tend to be more stable over time in the normal gut microbiota (14, 15). Furthermore, the use of Whole Genome Sequencing (WGS)-based strain typing has revealed that genetically unstable microbial species are associated with various human diseases (16). Nevertheless, stable genomic features of a given microbiome, such as single nucleotide polymorphisms (SNPs) and structural variations (SVs), may serve as a host-specific fingerprint of the microbiome, potentially enabling the identification of individuals (14). These findings suggest that a comprehensive understanding of the normal variations in the gut microbiota across the lifespan and detailed strain-level analyses would facilitate the identification of “true disease-associated microbial signatures” and enable personalized treatment options.

FACTORS AFFECTING THE GUT MICROBIOME

Accumulating evidence shows that an alteration of the gut microbiome is not only a fingerprint that can track individual health conditions, but also its association and causality with various human diseases have been suggested (10). Consequently, there are many ongoing studies to find and understand factors that induce changes in the gut microbiome.

Lifestyle

Lifestyle factors can significantly impact the composition and function of the human gut microbiome. Among them, diet is a key determinant as it can regulate the microbial metabolic processes depending on the specific macronutrients and ingredients in a diet (17). For example, Mediterranean diets, which are high in fiber, monounsaturated fat and antioxidants, can lead to changes in the structure and metabolomic profile of the gut microbiome, resulting in higher levels of fecal short-chain fatty acids (SCFAs) and a reduced risk of obesity and cardiovascular diseases. Similarly, the Hadza hunter-gatherers who consume a diet rich in complex carbohydrates, have increased diversity of the gut microbiota (18). On the other hand, long-term consumption of Western diets or diets deficient in microbiota-accessible carbohydrates found in dietary fiber can lead to the extinction of several gut microbiota taxa over generations, emphasizing the role of diet as a driving force in microbiota composition and function, and consequently our health and disease (19).

Geographical differences in gut microbiome composition may also be related to dietary differences. The migration to the United States from non-western countries, which have diets dominated by maize and cassava, to the Western diet, which is high in sugar, fat, and protein, resulted in an immediate loss of gut microbiome diversity and function (20). Similarities in microbial communities between the U.S. control group and second-generation immigrants indicate that dietary changes may influence microbial community shifts, but other factors such as stress, exercise, antibiotics and antiparasitic treatment may play a role as well (20).

In addition to diet, other lifestyle factors such as smoking, alcohol consumption and exercise have been shown to impact the gut microbiome. Smoking and alcohol consumption reduce gut microbial diversity, increase intestinal permeability and decrease the production of SCFAs (21). Interestingly, smoking caused more severe gut dysbiosis than alcohol consumption, but the combination of smoking and alcohol does not worsen it compared to either group alone (21), suggesting that the interaction effect of tobacco and alcohol on the gut microbiota is complex. Moderate-intensity endurance exercise has been shown to reduce inflammation, improve body composition, contribute to the diversity and composition of gut microbiota, and have effects on metabolic contributions to human health. Conversely, irregular and exhausting training can increase intestinal barrier permeability, decrease intestinal mucus thickness, and may contribute to gut dysbiosis (22).

Individual physiological patterns, such as stool frequency, correlate with gut microbial richness, distribution, and composition (23). Taken together, lifestyle factors can have wide-ranging effects on human health by modulating the gut microbiome. Therefore, it is important to identify and understand the factors that influence the development of microbial communities to promote better health outcomes.

Medication

Orally administered medications reach the intestine, where they may interact with the gut microbiome, potentially affecting the effectiveness of the drugs (24). Notably, drugs designed to target human cells, not microbes, have also been shown to alter the composition and function of the gut microbiome (25). For example, metformin, a commonly used medication for type 2 diabetes, has been found to alter the gut microbiome composition in mice and humans, showing a positive correlation with the abundance of Escherichia and Akkermansia (26). Furthermore, the metformin-altered human microbiota or A. muciniphila improved glucose tolerance in mice, indicating that the altered gut microbiota plays a role in some of metformin’s antidiabetic effects (27). Similarly, rosuvastatin, a commonly used cholesterol-lowering drug of the statin class, has been shown to restore the altered gut microbiota induced by a high-fat diet. The composition of the gut microbiota can also affect the bioavailability of statins by modulating the metabolism of bile acids (28). Captopril, an antihypertensive drug, has been shown to increase the amounts of bacterial spores such as Parabacteroides, Mucispirillum and Allobaculum and induce long-lasting antihypertensive effects (29).

The gut microbiome can also, directly and indirectly, influence an individual’s response to a drug and immunotherapy in cancer treatment by enzymatically transforming the drug’s structure and altering its bioavailability, bioactivity, or toxicity. For example, digoxin, a cardiac glycoside, can be metabolized to cardio-inactive dihydrodigoxin by Eggerthella lenta, and brivudine, an antiviral drug, can be metabolized by Bacteroides, to a potentially toxic metabolite (30, 31). In addition, the gut microbiota can metabolize levodopa, a medication for Parkinson’s disease management, potentially reducing drug bioavailability. In addition, Enterococcus faecalis or E. lenta converts levodopa to m-tyramine leading to side effects (32).

The gut microbiome’s role in drug metabolism and efficacy underscores the importance of investigating the bidirectional interactions between the gut microbiome and drugs. Additionally, given that the human gut microbiota is highly individualized, it is speculated that the gut microbiota is one of the primary variables determining drug effectiveness in individuals (31).

Antibiotics

Antibiotics can disrupt the balance of the gut microbiota by increasing pathogenic bacteria including Enterobacteriaceae and Fusobacterium nucleatum, while reducing beneficial bacteria such as Bifidobacterium and butyrate producers, including Faecalibacterium prausnitzii and Eubacterium spp. (33). This disruption can have significant impacts on the host, affecting immune regulation, metabolic activities and overall health (34). Antibiotic-induced changes in the gut microbiota can have long-term effects on community structure, and individuals may have different recovery patterns (35). Additionally, the metabolic changes caused by antibiotic-induced alterations in the gut microbiota can contribute to various chronic and neurological diseases (36).

The human gut microbiota is a reservoir for antibiotic resistance genes (ARGs) and resistant microorganisms. The current knowledge on the “resistome” and “antibiotic resistance potential” suggests that the rapid development and dissemination of ARGs is largely due to antibiotic abuse and misuse, and bacteria carrying antibiotic resistance can persist in the human gut for years even after short-term antibiotic treatment (37).

Recent studies have investigated the use of the gut microbiome to restore dysbiosis caused by antibiotic use. For example, an engineered strain of Lactococcus lactis has been developed to degrade β-lactam antibiotics and alleviate their negative effects on commensal gut bacteria (38). Additionally, probiotics such as Bacillus clausii have shown to be promising in preventing antibiotic-associated diarrhea and improving Helicobacter pylori eradication rates (39). Fecal microbiota transplantation (FMT) is now widely used to treat antibiotic-induced gut dysbiosis, particularly in cases of Clostridium difficile and vancomycin-resistant E. faecium infections in both human cohorts and mouse models (40). The FDA recently approved SER-109, known as VOWST, oral capsules, composed of live and purified Firmicutes bacterial spores derived from healthy donors, for the prevention of recurrent C. difficile infection. This follows an earlier approval of REBYOTA, a microbiota suspension for rectal administration aiming at restoring the gut microbiota (41). These developments highlight the potential of microbiota-based live biotherapeutic products. Nevertheless, further research is needed to determine the optimal formulation and dosage, and ensure clinical safety and efficacy.

LIFE-LONG EFFECTS OF ANTIBIOTICS-INDUCED MICROBIOME DISRUPTION IN THE EARLY LIFE

Epidemiological studies increasingly suggest that exposure to biological and psychosocial hazards during the first 3 years of life can have long-term effects on overall health, including cardiovascular, immune, metabolic and brain health (42). The colonization of the gut microbiota during this period is critical for long-term health outcomes (2).

Especially, exposure to antibitics affects the gut microbiome in early life. Notably, antibiotics are the most frequently prescribed medication to infants. Exposure to antibiotics in early life can lead to a rapid decrease in the diversity of the gut microbiome through a transient disruption. While this can recover over time, such changes can have long-lasting consequences on the immune and metabolic programming of infants, and may contribute to the development of chronic diseases later in life (6).

The use of antibiotics during pregnancy can affect the maternal gut microbiome, consequently influencing the transmission of microbiota from the mother to the newborn and potentially leading to various health conditions, including childhood atopy, asthma, allergies and obesity (43). Observational studies in humans have shown that antibiotics used during the second and third trimesters of pregnancy are associated with an increased risk of obesity, allergy and asthma in offsprings (43). In murine models, the administration of a low dose of penicillin during late pregnancy has been shown to alter the microbiota composition of the offspring, leading to metabolic changes associated with obesity, diabetes and non-alcoholic fatty liver disease (44). Interestingly, exposure to vancomycin during the prenatal period in neonatal mice models increased susceptibility to allergic asthma, while the same antibiotic exposure in adulthood had no such effect (45). Moreover, prenatal antibiotics exposure in murine models altered the gut microbiome profiles of offspring, including bacterial metabolic pathways, and influenced the immune function of post-weaned prepubescent offspring (46). The effects of antibiotic use during pregnancy varies across different types of antibiotics, with narrow-spectrum antibiotics such as pivmecillinam, erythromycin and clindamycin showing no association with overweight in offspring, while some broad-spectrum antibiotics such as ampicillin, amoxicillin and sulfamethizole may increase the risk of infant obesity in humans (47). Antibiotic use during the first year of life has also been associated with a higher risk of childhood obesity, pneumonia, asthma, allergies and related symptoms (48, 49).

Tracking antibiotic-exposed mice from infancy to old age revealed that, despite receiving the same antibiotics, distinct and functionally diverse changes in the microbial communities occur at different stages of life, suggesting that there is a critical period in life where antibiotic exposure has long-lasting effects on metabolism, immunity and lifespan (50). Indeed, antibiotic exposure in early life delays the development of the gut microbial community, with the most significant impact occurring between 6 and 12 months of age (5). This delay has been linked to the accelerated onset of childhood overweight and obesity between the ages of 1 and 2 and a half years in humans (49). In murine models, the early-life administration of antibiotics such as low-dose penicillin, tylosin and a mixture of amoxicillin and tylosin induced a significant increase in total and fat mass due to metabolic changes, demonstrated by the transfer of antibiotic-induced altered microbiome to germ-free mice (6, 51). Short-term antibiotic treatment, for less than 3 days, in infants has only a temporary and mild effect on the gut microbial composition and metabolites, while intensive antibiotic exposure can lead to a continuous decrease in the diversity of the microbiome (52). Interestingly, a recent study using rats suggests that selective reshaping of the gut microbiota through the use of appropriate antibiotics during the gestation and lactation period may have long-term benefits against pediatric hypertension (53). Therefore, it is important to use appropriate antibiotic combinations, doses and timing; this helps to minimize the risks associated with inevitable antibiotic exposure and achieve optimum therapeutic effects while promoting the rapid recovery of the gut microbial homeostasis.

FACTORS AFFECTING THE ESTABLISHMENT OF THE EARLY-LIFE MICROBIOME

The establishment and maturation of the gut microbiome in infants can vary, and microbiome perturbations during the first weeks to months of life can affect infant growth and health (54). The establishment of the gut microbiota during infancy is influenced by multiple factors, such as prenatal, peripartum and environmental factors (Fig. 1) (55). Given its close and causal relationship with later-life quality, we will discuss factors affecting the establishment of the early microbiome.

Maternal effect during pregnancy

Environmental factors such as diet, drug intake and stress can alter the maternal microbiome during pregnancy, consequently influencing the intrauterine environment and significantly affecting the fetal and infant microbiome (56-58). Maternal transmission of gut bacteria provides a microbial ‘starter kit’ for infants, promoting healthy growth and disease resistance (59). Microbial strains from multiple maternal body sites are transferred to the infant microbiome (4, 60). Metagenomic profiles have shown that maternal gut strains are more persistent in the infant’s gut, while maternal skin and vaginal strains colonize only transiently (60).

Metagenomics analysis has reported that excessive gestational weight gain in mothers (BMI > 25) alters the abundance and diversity of the gut microbiome, leading to a significant reduction in metabolic signaling and energy regulation-related bacterial communities, including Enterococcus, Acinetobacter and Pseudomonas (61). Moreover, research involving birth cohorts and murine models has demonstrated that the offspring born to mothers who consumed a high-fat diet or were overweight during pregnancy are enriched with Firmicutes and have impaired insulin sensitivity. These offsprings have a 5-fold higher risk of being overweight and developing metabolic diseases compared to those born to mothers of normal weight (62). Additionally, low-fiber diets during lactation can result in lasting microbiota dysbiosis in offsprings, characterized by reduced taxonomic diversity, increased abundance of Proteobacteria and increased adiposity when exposed to an obesogenic diet (56). Antibiotic intake during pregnancy can also disrupt the transmission of maternal-derived pioneer gut bacteria during birth, potentially affecting metabolic health. In a study involving pregnant spontaneously hypertensive rats treated with captopril water, changes in the gut microbiota were observed, and some of the changes were reflected in their male offsprings that showed persistently decreased systolic blood pressure and improvements in gut inflammation and permeability (57).

Maternal prenatal psychosocial stress is another factor that influences the long-term development of the infant microbiome (58). Continuous exposure to stress has been reported to disturb the vaginal bacterial structure and composition, which can be transmitted to the offsprings, affecting their gut microbiome composition and neurodevelopment (63). Prenatal depression, which affects 10-20% of pregnant women, can have various effects on the infant’s health, including behavior problems and increased susceptibility to physical illnesses (64). Infants born to mothers with high levels of depression and anxiety showed a reduction in the abundance of beneficial bacteria such as Bifidobacterium, and Lactobacillus, which have been shown to have a role in the gut-brain axis and immune modulation (65). For example, depression-induced maternal pro-inflammatory cytokines can promote dysfunctional neurodevelopment in offspring (65, 66).

Prenatal exposure to a smoking environment also greatly affects the composition of the gut microbiome in infants. The majority (78%) of infants born to mothers who smoked were born prematurely and infants from non-smoking families showed higher alpha diversity of gut microbiota compared to infants from smoking families. This suggests that secondhand smoke can affect the intestinal microbial community of neonates (67). Additionally, maternal smoking is associated with low birth weight in children and can also increase the abundance of Firmicutes during the first 3 months of life, thereby increasing the risk of obesity in children between the ages of 1 and 3 years (68).

Gestational age

The microbiota in infants undergoes dynamic changes in composition, and the temporal colonization pattern of the intestinal microbiota during the early stages of life may have an important contribution to long-term health (2, 69). However, most studies investigating the development of the gut microbiome in early life have primarily focused on full-term infants. In recent years, the prevalence of premature birth has increased, with approximately 5-18% of live births in 2019 classified as preterm (babies born before 37 weeks of gestational age) (70). Importantly, gestational age is considered a significant factor contributing to the development and maturation of the gut microbiome during early life (71).

Recent analyses of gut microbiota in infants have revealed significant differences in the meconium and stool between full-term and preterm infants (refer to Table 1). Although there were no significant differences in alpha diversity, the beta diversity analysis indicated distinct structures of the gut microbiota in each group (72). The most abundant bacteria at the phylum level in each group were Proteobacteria, Firmicutes and Bacteroidetes (72, 73).

Especially, there is a prediction that preterm infants may be colonized by potentially pathogenic facultative anaerobic bacteria such as Enterobacter, Escherichia and Klebsiella, with delayed colonization of commensal strict anaerobes such as Bifidobacterium, Bacteroides and Clostridium (74-77).

Preterm infants are predominantly born via cesarean section (CS), resulting in a gut microbiome that is more commonly colonized by the skin and environmental bacteria rather than the vaginal and rectal microbiome (78). Also, exposure to the neonatal intensive care unit, including sterile incubator nursing and broad-spectrum antibiotics, limits the acquisition and development of normal microbiota. This can lead to altered levels of Clostridium and B. fragilis, resulting in a different gut microbiome compared to full-term infants (74, 79). Antibiotic exposure in preterm infants increases the presence of antibiotic resistance genes or abundance of multi-drug resistant members of Klebsiella, Escherichia, Enterobacter and Enterococcus genera (80-82).

The delays and alterations in the colonization of commensal bacteria in the gut of preterm infants have been associated with the development of metabolic and gastrointestinal illnesses such as Type 1 diabetes, asthma and allergies, necrotizing enterocolitis (NEC) and late-onset sepsis (LOS), which leads to atopy and neurodevelopmental effects in childhood and later life (71, 83-86). For example, early Bifidobacterium deficiency in preterm infants seems to be a negative biomarker of adverse neurological outcomes and NEC (77, 87). Moreover, the transplantation of meconium microbiota from preterm infants into germ-free mice resulted in growth disorders, impaired intestinal immune function and metabolic abnormalities (88).

Current research provides evidence that premature birth impacts infant growth and development. However, our understanding of the gut microbiota in preterm infants from infancy to childhood is still limited. Future research is needed to identify the critical period during which interventions can be implemented to normalize infant growth, development and the microbiome. This knowledge would be invaluable for improving health outcomes in preterm infants.

Delivery mode

The delivery mode is a common major factor that can directly affect early microbial colonization (5). Vaginally delivered infants are enriched with bacteria such as Lactobacillus, Prevotella and Sneathia, which are present in the mother’s intestine and vagina. In particular, bacteria derived from the mother’s intestine persist more in the infant’s intestine (5, 60). However, infants born via CS are colonized with bacteria similar to those found on the mother’s skin and oral cavity since vertical transmission is blocked (78). This results in relatively reduced alpha diversity and lower levels of Bifidobacterium and Bacteroides, and higher levels of Clostridium, Lactobacillus, Enterobacter, Enterococcus, and Staphylococcus (84, 89). Within the first week after birth, there was no difference in the gut microbiota between vaginally delivered infants and CS-born infants (90). However, in the second week, Bacteroides in CS-born infants disappeared due to the absence of Bacteroides supporting species (4, 90). Furthermore, CS-born infants have lower alpha diversity during the first year (5, 91). Nevertheless, this difference normalizes in 3 to 5 years as the gut microbiota continued to mature (91). In contrast to term infants, the birth mode does not significantly impact microbial characteristics in preterm infants (92). This may be attributed to the dominant influence of neonatal intensive care unit (NICU) exposures, such as early antibiotic use and the absence of Bacteroides colonization in preterm infants (92).

Infant feeding type

Numerous studies have demonstrated that infant feeding type significantly influences the composition, development, and diversity of early microbiomes, which has a significant impact on immune function. Human breast milk considered the “gold standard” of infant nutrition, contains a wide spectrum of human milk microbiota that plays a crucial role in gastrointestinal colonization in infants. These microbiota include Firmicutes (i.e., Streptococcus, Staphylococcus, Lactobacillus), Proteobacteria (i.e., Serratia, Pseudomonas, Ralstonia, Sphingomonas), and Actinobacteria (i.e., Propionibacteria, Corynebacteria, Bifidobacterium) (93, 94). In addition to fulfilling the infant’s initial nutrition requirements, breast milk contains immunoglobulins, fatty acids, hormones and cytokines that contribute to the infant’s health and development (93). Furthermore, breastfeeding can play a vital role in enhancing gut microbiome diversity by selectively promoting the growth of beneficial bacteria such as Bifidobacterium and Lactobacillus species, which can break down specific oligosaccharides present in human milk (94). Consequently, the gut microbiome of breast-fed infants is typically dominated by Bifidobacterium and Lactobacillus species, which are associated with a lower risk of developing allergic diseases (78).

Infant formula milk formulation, including cow-milk-based formula and soy-based formula, mimics the nutritional composition of breast milk (95). However, cow’s milk-based formula contains higher levels of fat, minerals and proteins compared to human breast milk (96). Recent studies have shown that high protein content in infant formula is associated with excess weight gain in infants, which can lead to a 20% risk of being overweight later in life and an increased risk of food allergies (78, 97). Formula feeding is associated with a higher incidence of alterations in the infant gut microbiota compared to breast-fed infants such as in favor of proinflammatory taxa, increased gut permeability, and antibiotic resistance load (98). Despite efforts by formula manufacturers to replicate nutrients similar to those in human milk, the gut microbiota of breast-fed and formula-fed infants remain distinct (99).

In the relationship between delivery mode and infant feeding type, it has been observed that there was no difference in the abundance of Bifidobacterium according to feeding type in infants delivered vaginally. However, in CS-born infants, breast-fed infants have higher levels of B. longum, B. breve, B. bifidum, and B. pseudocatenulatum compared to formula milk-fed infants (100, 101). Interestingly, a recent report suggests that CS-born infants who do not have direct exposure to the birth canal may compensate for the transmission of the maternal microbiome through other routes, such as breast milk microbiota (4). Also, despite the high risk for dysbiosis of the gut microbiome in preterm babies, mother’s own milk has been shown to contribute to the development of a diverse and balanced microbial community in early life compared to donated human milk and formula milk (102).

Nevertheless, regardless of the delivery mode and infant feeding type, the introduction of solid food at around 1 year after birth leads to a more rapid and stable maturation of the gut microbiome, and an increase in diversity (100). As a result, by the age of 5 years, there is minimal difference in the initial gut microbial composition of infants based on the delivery method (91). However, even with the normalization of the microbiome, there may still be long-lasting effects from the previous microbiome associated with delivery modes, which could manifest in differences in disease prevalence (Fig. 1) (103).

EFFORTS TO RESTORE EARLY MICROBIOME

The development of the gut microbiome in the early stages of life can be compromised in infants born via CS, preterm births, or exposure to antibiotics (5, 82, 104). Currently, commonly used intervention strategies to restore developmental impairment of the early gut microbiome in neonates include fecal microbiota transplantation (FMT), probiotics and nutrients (Fig. 2) (104-106). When maternal vaginal microbes were transplanted orally to CS-born infants, it did not show significant changes in gut microbiome composition and functional potential (107). Meanwhile, when CS-born infants immediately received a swab of the maternal vaginal microbiota, starting from the mouth, then the face and the rest of the body, a partial recovery of gut, oral and skin microbiota occurred similar to infants born vaginally, during the first 30 days (108). However, regardless of administration techniques, vaginal seeding has not sufficiently restored Bacteroides colonization and abundance in the gut microbiota of CS-born infants (108). Conversely, orally delivered maternal FMT exhibits a composition of the gut microbiota similar to that observed in infants born vaginally, indicating that the maternal fecal microbiota may be a more effective method for restoring Bacteroides levels in CS-born infants (104, 107). The restorative effect of FMT has also been observed in animal models, which affected the composition of the gut microbiota, prevented mucosal damage and NEC in a preterm pig model, and effectively reversed NEC damage in a preterm mouse model (109, 110). However, there are still risks associated with introducing living microorganisms, particularly the potential for disease occurrence due to the weakened immune system of the recipient (111). Therefore, as for safety, the importance of establishing donor screening criteria has been proposed. For example, current standard processes for the most successful and frequently performed treatment of C. difficile infection with FMT typically involve pre-screening with a medical history evaluation and blood and stool tests to detect the presence of infectious diseases, but the efficiency of screening is low and the focus is mainly on excluding infections and underlying diseases (112). Therefore, it is necessary to accumulate data on the occurrence of complications through longitudinal studies and understand how environmental factors or personal characteristics affect FMT clinical responses (112).

The supplementation of probiotics to preterm infants has been reported to be effective in preventing sepsis and major disorders such as NEC (106). Probiotic supplementation, containing species of Bifidobacterium, Lactobacillus and Streptococcus, in preterm infants and extremely preterm infants less than 5 days old, has been shown to increase the abundance of probiotic species in the gut microbiota of preterm infants, and effectively prevent serious morbidities associated with prematurity such as NEC and LOS (113). In addition, longitudinal analysis has demonstrated that the dominance of B. breve through probiotic supplementation is associated with accelerated maturation of the preterm infant gut. These findings suggest that the strain-specific effect is an important aspect to consider when designing an early intervention targeting the gut microbiome in preterm infants (114).

Breast and formula milk are the major sources of nutrition for newborns and can be used as strategies for nutritional supplementation (115). Human milk oligosaccharides (HMOs) are complex and unconjugated sugar structures in human milk, which are proposed to support infant growth, development and health. HMOs may have a protective impact on NEC in preterm infants by modulating the immune system and gut microbiota (105, 116). Furthermore, the addition of 2 HMOs (2’-fucosyllactose and lacto-N-neotetraose) in commercial formulas for term infants can produce gut microbiome compositions and functions, similar to those of breast-fed infants (78, 117). HMOs can promote the growth of Bifidobacterium and can be utilized by Akkermansia, suggesting the potential of enhancing the intestinal barrier function and immune system by guiding the healthy microbiota (105, 118). While various early intervention strategies have been proposed, further research is needed to understand the precise mechanisms for restoring disrupted or immature gut microbiota. It is crucial to establish reliable and effective strategies that ensure stability.

PERSPECTIVE

The gut microbiome and its metabolic processes are dynamic systems that can be easily altered by various factors. Antibiotic exposure often leads to the bloom of pathogens such as Enterococcus and Fusobacterium and the depletion of beneficial Bifidobacterium species, but most of the common microbiome composition is restored within 1.5 months after antibiotic exposure (119). However, in the long term, there may be “antibiotic scarring”, characterized by altered metabolic output and an increased antibiotic resistance burden even after the administration of antibiotics ceases (33). Persistent metagenomic signatures, such as enriched gut antibiotic resistome and carriage of multidrug-resistant Enterobacteriaceae, may remain and contribute to chronic pathology despite the recovery of the gut microbiota (120).

The early life period is particularly critical for host-microbial metabolic interactions, and altered metabolic phenotypes driven by delivery mode, infant feeding type and antibiotic exposure can persist and affect the long-term risk of chronic disease, even if early gut microbial perturbations recover (6, 121). Therefore, further research is needed to identify critical periods during which restoring the microbiome composition can lead to phenotype recovery.

The rising global preterm birth rates have emphasized the need to comprehensively investigate the complex microbial environment within the human gut using a combination of metagenomics and metabolomics approaches for a deeper understanding of maternal factors associated with infant health from the fetal stage (101). From a metagenomics perspective, taking probiotics during early pregnancy is an alternative treatment for preventing bacterial vaginosis-associated preterm birth and spontaneous preterm birth (122). In addition, starting supplementation of a probiotic mixture or human milk fortifier within a month after birth can have beneficial effects on the gut microbiome maturation as a critical developmental window of preterm infants, consequently improving clinical outcomes (123, 124).

From a metabolic perspective, the maternal gut microbiota can affect not only the maternal compartment but also the fetus in the sterile uterus through the modulation of microbial metabolites (3, 125). The microbial metabolites may modulate the infant gut microbial ecosystem, immunity and long-term metabolic effects on the offspring, including increased adiposity, glucose intolerance and neural development in adulthood (43, 126). Recently, the discovery of beneficial effects on infants through the release of indole-3-lactic acid and γ-linolenic acid via breast milk metabolization offers potential therapeutic applications, highlighting the possibility of harnessing the transmission of microbial metabolites from mother to infant for therapeutic purposes (127). In addition, as dynamic metabolic changes occur during pregnancy, potential biomarkers of gestational age prediction and preterm birth have been observed in blood and vaginal samples (128). For example, maternal metabolites can be used as early biomarkers for preterm birth which provides a basis for prevention and treatment. Moreover, considering the potential variation in transplantation outcomes based on subtle species-level donor differences (123), the use of validated metabolites that are more stable and reproducible than FMT can be considered as an alternative approach for gut microbiota restoration. It is also important to set the precise time point and dose of the early intervention to achieve the optimal efficacy and safety for the recovery of the early gut microbiota.

ACKNOWLEDGEMENTS
This paper is supported by the Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korean government (the Ministry of Science and ICT, the Ministry of Health & Welfare) (KFRM 22A0301L1), the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. 2023R1A2C1002876), and the Basic Science Institute (National Research Facilities and Equipment Center) grant funded by the Ministry of Education (No. 2021R1A6C101A390). Additionally, this work is supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2021R1A6A3A01086599 to N.Y.P).
CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. The significant impact of the initial formation and development of early-life gut microbiota on later-life disease development. Newborns undergo temporal colonization of the microbiota based on gestational age. Delivery mode (vaginal birth vs. cesarean section), feeding type (breast-fed vs. formula-fed), and antibiotic intake influence the establishment and maturation of the gut microbiota during the first weeks to months of life. Although differences in early gut microbiota normalize after 3-5 years, the metabolic phenotype persists, contributing to long-term risks of chronic diseases such as neurological diseases, obesity, type 2 diabetes, and non-alcoholic fatty liver disease. However, the understanding of the influence of gut microbiome on long-term health in preterm infants is still limited, designated as a question mark in the figure. PD, Parkinson’s disease; AD, Alzheimer’s disease. Fig. 1 created using www.biorender.com.
Fig. 2. Early interventions to achieve microbial recovery in infants with disrupted gut microbiota. The first 3 years from birth are considered a critical window for early intervention from gut microbial disruption by premature birth, antibiotics exposure, and cesarean section delivery, which can influence long-term health outcomes. Current efforts focus on fecal and vaginal microbiota transplantation from the mother to infants, as well as nutritional supplementation (i.e., human milk oligosaccharides), and probiotics, as potential strategies to restore developmental impairments in the early gut microbiota. Additionally, microbial metabolite would constitute another potential therapeutic option with improved efficacy and safety compared to live microbiota transplantation.
TABLE

Alteration of the early-life colonization of the human gut microbiota in compliance with the gestational age (GA)

Preterm (GA) Control group Sample numbers (type) Microbiota Change Microbiota analysis (method) References
Full term (36-40 weeks) Mother, father, siblings 14 (feces) Flexibacter-Cytophaga-Bacteroides, Proteobacteria, Gram-positive Bacteria (Firmicutes and Actinobacteria) Increase 16S rRNA (Microarrays) (69)
Late preterm infants (34-36 weeks) Full-term infants 43 (feces) Clostridium perfringens Increase 16S rRNA (qPCR) (74)
Akkermansia, Bifidobacterium Decrease
Preterm infants with NEC, LOS (< 33 weeks) Infants with no infection 24 (feces) Enterococcus, Streptococcus, Peptoclostridium (NEC), Klebsiella (LOS) Increase 16S rRNA (Miseq) (80)
Preterm infants (32-37 weeks) Full-term infants 57 (vaginal) Lactobacillus crispatus, Lactobacillus iners Decrease Shotgun metagenomic sequencing (Nextseq) (75)
Preterm infants (< 32 weeks) Breast milk-fed preterm-infants 27 (feces) Enterobacteriaceae, Enterococcaceae, Staphylococcaceae Increase 16S rRNA (DGGE) (76)
Preterm infants with LOS (< 32 weeks) Infants with no infection 28 (feces) Proteobacteria, Actinobacteria Increase 16S rRNA (DGGE) (77)
Bifidobacterium, Firmicutes, Bacteroidetes Decrease
Preterm infants (≤ 32 weeks) Non-used diaper 14 (feces, meconium) Bacilli, Firmicutes (meconium), Proteobacteria (feces) Increase 16S rRNA (DGGE, HITChip) (72)
Preterm infants (30-35 weeks) Breast milk-fed full-term infants 41 (feces) Facultative anaerobic microorganisms (Enterococcaceae, Enterobacteriaceae, Weissella, Klebsiella pneumoniae) Increase 16S rRNA (qPCR) (79)
Strict anaerobes (Bifidobacterium, Bacteroides, Atopobium) Decrease
Extremely preterm infants (< 28, 28-32 weeks) Full-term infants (37-42 weeks) 70 (meconium) Klebsiella, Staphylococcus, Sphingomonas Increase 16S rRNA (Hiseq) (71)
Very low birth weight (VLBW) (27.9 ± 2.2 weeks) Same infants at 29.8 ± 2.3, 31.2 ± 1.9, and 32.6 ± 1.9 weeks postnatal age 45 (feces) Firmicutes, Bacilli Increase 16S rRNA (Miseq) (81)
Gammaproteobacteria, Actinobacteria, Clostridia Decrease
Preterm infants with NEC/LOS (27 weeks) Healthy infants 38 (feces) Enterobacter, Staphylococcus Increase 16S rRNA (DGGE) (86)
Extremely preterm infants (25.1-31.2 weeks) The same infants after birth 45 (feces 60 days postnatal age) Enterococcus., Enterobacter, Staphylococcus Increase 16S rRNA (MiSeq) (82)
Bifidobacterium Decrease
Preterm infants (24-32 weeks) The same infants 2 years after birth 30 (meconium, feces) Proteobacteria, Fusobacteria Increase 16S rRNA (HITChip) (73)
Firmicutes, Bacteroidetes Decrease

NEC, necrotizing enterocolitis; LOS, late-onset sepsis; qPCR, quantitative PCR; DGGE, denaturing gradient gel electrophoresis; HITChip, Human intestinal tract chip.


REFERENCES
  1. Koh A and Bäckhed F (2020) From association to causality: the role of the gut microbiota and its functional products on host metabolism. Mol Cell 78, 584-596.
    Pubmed CrossRef
  2. Sarkar A, Yoo JY, Valeria Ozorio Dutra S, Morgan KH and Groer M (2021) The association between early-life gut microbiota and long-term health and diseases. J Clin Med 10, 459.
    Pubmed KoreaMed CrossRef
  3. Pessa-Morikawa T, Husso A and Kärkkäinen O et al (2022) Maternal microbiota-derived metabolic profile in fetal murine intestine, brain and placenta. BMC Microbiol 22, 46.
    Pubmed KoreaMed CrossRef
  4. Bogaert D, Van Beveren GJ and de Koff EM et al (2023) Mother-to-infant microbiota transmission and infant microbiota development across multiple body sites. Cell Host Microbe 31, 447-460.
    Pubmed CrossRef
  5. Bokulich NA, Chung J and Battaglia T et al (2016) Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 8, 343ra82.
    Pubmed KoreaMed CrossRef
  6. Cox LM, Yamanishi S and Sohn J et al (2014) Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158, 705-721.
    Pubmed KoreaMed CrossRef
  7. Sender R, Fuchs S and Milo R (2016) Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 14, e1002533.
    Pubmed KoreaMed CrossRef
  8. Cao H, Xu M and Dong W et al (2017) Secondary bile acid‐induced dysbiosis promotes intestinal carcinogenesis. Int J Cancer 140, 2545-2556.
    Pubmed CrossRef
  9. Ryu G, Kim H and Koh A (2021) Approaching precision medicine by tailoring the microbiota. Mamm Genome 32, 206-222.
    Pubmed CrossRef
  10. Chen Y, Zhou J and Wang L (2021) Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol 11, 625913.
    Pubmed KoreaMed CrossRef
  11. Zaneveld JR, McMinds R and Vega Thurber R (2017) Stress and stability: applying the Anna Karenina principle to animal microbiomes. Nat Microbiol 2, 17121.
    Pubmed CrossRef
  12. Watanabe S, Kameoka S and Shinozaki NO et al (2021) A cross-sectional analysis from the Mykinso Cohort Study: establishing reference ranges for Japanese gut microbial indices. Biosci Microbiota Food Health 40, 123-134.
    Pubmed KoreaMed CrossRef
  13. Ghosh TS, Das M, Jeffery IB and O'Toole PW (2020) Adjusting for age improves identification of gut microbiome alterations in multiple diseases. Elife 9, e50240.
    Pubmed KoreaMed CrossRef
  14. Chen L, Wang D and Garmaeva S et al (2021) The long-term genetic stability and individual specificity of the human gut microbiome. Cell 184, 2302-2315.
    Pubmed CrossRef
  15. Olsson LM, Boulund F and Nilsson S et al (2022) Dynamics of the normal gut microbiota: a longitudinal one-year population study in Sweden. Cell Host Microbe 30, 726-739.
    Pubmed CrossRef
  16. Vich Vila A, Imhann F and Collij V et al (2018) Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci Transl Med 10, eaap8914.
    Pubmed CrossRef
  17. Bourdeau-Julien I, Castonguay-Paradis S and Rochefort G et al (2023) The diet rapidly and differentially affects the gut microbiota and host lipid mediators in a healthy population. Microbiome 11, 1-16.
    Pubmed KoreaMed CrossRef
  18. Fragiadakis GK, Smits SA and Sonnenburg ED et al (2019) Links between environment, diet, and the hunter-gatherer microbiome. Gut Microbes 10, 216-227.
    Pubmed KoreaMed CrossRef
  19. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS and Sonnenburg JL (2016) Diet-induced extinctions in the gut microbiota compound over generations. Nature 529, 212-215.
    Pubmed KoreaMed CrossRef
  20. Vangay P, Johnson AJ and Ward TL et al (2018) US immigration westernizes the human gut microbiome. Cell 175, 962-972.
    Pubmed KoreaMed CrossRef
  21. Lin R, Zhang Y and Chen L et al (2020) The effects of cigarettes and alcohol on intestinal microbiota in healthy men. J Microbiol 58, 926-937.
    Pubmed CrossRef
  22. Wegierska AE, Charitos IA, Topi S, Potenza MA, Montagnani M and Santacroce L (2022) The connection between physical exercise and gut microbiota: implications for competitive sports athletes. Sports Med 52, 2355-2369.
    Pubmed KoreaMed CrossRef
  23. Kwon HJ, Lim JH, Kang D, Lim S, Park SJ and Kim JH (2019) Is stool frequency associated with the richness and community composition of gut microbiota?. Intest Res 17, 419-426.
    Pubmed KoreaMed CrossRef
  24. Weersma RK, Zhernakova A and Fu J (2020) Interaction between drugs and the gut microbiome. Gut 69, 1510-1519.
    Pubmed KoreaMed CrossRef
  25. Maier L, Pruteanu M and Kuhn M et al (2018) Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 555, 623-628.
    Pubmed KoreaMed CrossRef
  26. Forslund K, Hildebrand F and Nielsen T et al (2015) Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528, 262-266.
    Pubmed KoreaMed CrossRef
  27. Wu H, Esteve E and Tremaroli V et al (2017) Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med 23, 850-858.
    Pubmed CrossRef
  28. Kim J, Lee H and An J et al (2019) Alterations in gut microbiota by statin therapy and possible intermediate effects on hyperglycemia and hyperlipidemia. Front Microbiol 10, 1947.
    Pubmed KoreaMed CrossRef
  29. Yang T, Aquino V and Lobaton GO et al (2019) Sustained captopril‐induced reduction in blood pressure is associated with alterations in gut‐brain axis in the spontaneously hypertensive rat. J Am Heart Assoc 8, e010721.
    Pubmed KoreaMed CrossRef
  30. Haiser HJ, Seim KL, Balskus EP and Turnbaugh PJ (2014) Mechanistic insight into digoxin inactivation by Eggerthella lenta augments our understanding of its pharmacokinetics. Gut Microbes 5, 233-238.
    Pubmed KoreaMed CrossRef
  31. Wan Y and Zuo T (2022) Interplays between drugs and the gut microbiome. Gastroenterol Rep 10 (Oxf), goac009.
    Pubmed KoreaMed CrossRef
  32. Maini Rekdal V, Bess EN, Bisanz JE, Turnbaugh PJ and Balskus EP (2019) Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science 364, eaau6323.
    Pubmed KoreaMed CrossRef
  33. Anthony WE, Wang B and Sukhum KV et al (2022) Acute and persistent effects of commonly used antibiotics on the gut microbiome and resistome in healthy adults. Cell Rep 39, 110649.
    Pubmed KoreaMed CrossRef
  34. Patangia DV, Anthony Ryan C, Dempsey E, Paul Ross R and Stanton C (2022) Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 11, e1260.
    Pubmed KoreaMed CrossRef
  35. Bonine NG, Berger A and Altincatal A et al (2019) Impact of delayed appropriate antibiotic therapy on patient outcomes by antibiotic resistance status from serious gram-negative bacterial infections. Am J Med Sci 357, 103-110.
    Pubmed CrossRef
  36. Leong KS, Derraik JG, Hofman PL and Cutfield WS (2018) Antibiotics, gut microbiome and obesity. Clin Endocrinol 88, 185-200.
    Pubmed CrossRef
  37. Feng J, Li B and Jiang X et al (2018) Antibiotic resistome in a large‐scale healthy human gut microbiota deciphered by metagenomic and network analyses. Environ Microbiol 20, 355-368.
    Pubmed CrossRef
  38. Cubillos-Ruiz A, Alcantar MA, Donghia NM, Cárdenas P, Avila-Pacheco J and Collins JJ (2022) An engineered live biotherapeutic for the prevention of antibiotic-induced dysbiosis. Nat Biomed Eng 6, 910-921.
    Pubmed CrossRef
  39. Lahiri K, Jadhav K, Gahlowt P, Najmuddin F and Padmashree Y (2015) Bacillus Clausii as an adjuvant therapy in acute childhood Diarrhoea. IOSR-JDMS 14, 74-76.
    CrossRef
  40. Drekonja D, Reich J and Gezahegn S et al (2015) Fecal microbiota transplantation for Clostridium difficile infection: a systematic review. Ann Intern Med 162, 630-638.
    Pubmed CrossRef
  41. Mullard A (2023) FDA approves second microbiome-based C. difficile therapy. Nat Rev Drug Discov 6, 436.
    Pubmed CrossRef
  42. Nelson CA, Bhutta ZA, Harris NB, Danese A and Samara M (2020) Adversity in childhood is linked to mental and physical health throughout life. BMJ 371, m3048.
    Pubmed KoreaMed CrossRef
  43. Milliken S, Allen RM and Lamont RF (2019) The role of antimicrobial treatment during pregnancy on the neonatal gut microbiome and the development of atopy, asthma, allergy and obesity in childhood. Expert Opin Drug Saf 18, 173-185.
    Pubmed CrossRef
  44. Leclercq S, Mian FM and Stanisz AM et al (2017) Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat Commun 8, 15062.
    Pubmed KoreaMed CrossRef
  45. Russell SL, Gold MJ and Hartmann M et al (2012) Early life antibiotic‐driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep 13, 440-447.
    Pubmed KoreaMed CrossRef
  46. Madany AM, Hughes HK and Ashwood P (2022) Prenatal maternal antibiotics treatment alters the gut microbiota and immune function of post-weaned prepubescent offspring. Int J Mol Sci 23, 12879.
    Pubmed KoreaMed CrossRef
  47. Jess T, Morgen CS and Harpsøe MC et al (2019) Antibiotic use during pregnancy and childhood overweight: a population-based nationwide cohort study. Sci Rep 9, 11528.
    Pubmed KoreaMed CrossRef
  48. Zou Z, Liu W, Huang C, Sun C and Zhang J (2020) First-year antibiotics exposure in relation to childhood asthma, allergies, and airway illnesses. Int J Environ Res Public Health 17, 5700.
    Pubmed KoreaMed CrossRef
  49. Li P, Chang X and Chen X et al (2022) Early-life antibiotic exposure increases the risk of childhood overweight and obesity in relation to dysbiosis of gut microbiota: a birth cohort study. Ann Clin Microbiol Antimicrob 21, 1-14.
    Pubmed KoreaMed CrossRef
  50. Lynn MA, Eden G and Ryan FJ et al (2021) The composition of the gut microbiota following early-life antibiotic exposure affects host health and longevity in later life. Cell Rep 36, 109564.
    Pubmed CrossRef
  51. Nobel YR, Cox LM and Kirigin FF et al (2015) Metabolic and metagenomic outcomes from early-life pulsed antibiotic treatment. Nat commun 6, 7486.
    Pubmed KoreaMed CrossRef
  52. Greenwood C, Morrow AL and Lagomarcino AJ et al (2014) Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J Pediatr 165, 23-29.
    Pubmed KoreaMed CrossRef
  53. Galla S, Chakraborty S and Cheng X et al (2020) Exposure to amoxicillin in early life is associated with changes in gut microbiota and reduction in blood pressure: findings from a study on rat dams and offspring. J Am Heart Assoc 9, e014373.
    Pubmed KoreaMed CrossRef
  54. Raju TN (2013) Moderately preterm, late preterm and early term infants: research needs. Clin Perinatol 40, 791-797.
    Pubmed KoreaMed CrossRef
  55. Vandenplas Y, Carnielli V and Ksiazyk J et al (2020) Factors affecting early-life intestinal microbiota development. Nutrition 78, 110812.
    Pubmed CrossRef
  56. Zou J, Ngo VL, Wang Y, Wang Y and Gewirtz AT (2023) Maternal fiber deprivation alters microbiota in offspring, resulting in low-grade inflammation and predisposition to obesity. Cell Host Microbe 31, 45-57.
    Pubmed CrossRef
  57. Li HB, Yang T, Richards EM, Pepine CJ and Raizada MK (2020) Maternal treatment with captopril persistently alters gut-brain communication and attenuates hypertension of male offspring. Hypertension 75, 1315-1324.
    Pubmed KoreaMed CrossRef
  58. Van den Bergh BR, van den Heuvel MI and Lahti M et al (2020) Prenatal developmental origins of behavior and mental health: the influence of maternal stress in pregnancy. Neurosci Biobehav Rev 117, 26-64.
    Pubmed CrossRef
  59. Browne HP, Shao Y and Lawley TD (2022) Mother-infant transmission of human microbiota. Curr Opin 69, 102173.
    Pubmed CrossRef
  60. Ferretti P, Pasolli E and Tett A et al (2018) Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host Microbe 24, 133-145.
    Pubmed KoreaMed CrossRef
  61. Collado MC, Laitinen K, Salminen S and Isolauri E (2012) Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr Res 72, 77-85.
    Pubmed CrossRef
  62. Tun HM, Bridgman SL and Chari R et al (2018) Roles of birth mode and infant gut microbiota in intergenerational transmission of overweight and obesity from mother to offspring. JAMA Pediatr 172, 368-377.
    Pubmed KoreaMed CrossRef
  63. Jašarević E, Howard CD, Misic AM, Beiting DP and Bale TL (2017) Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci Rep 7, 1-13.
    Pubmed KoreaMed CrossRef
  64. Rodriguez N, Tun HM, Field CJ, Mandhane PJ, Scott JA and Kozyrskyj AL (2021) Prenatal depression, breastfeeding, and infant gut microbiota. Front Microbiol 12, 664257.
    Pubmed KoreaMed CrossRef
  65. Galley JD, Mashburn-Warren L and Blalock LC et al (2023) Maternal anxiety, depression and stress affects offspring gut microbiome diversity and bifidobacterial abundances. Brain Behav Immun 107, 253-264.
    Pubmed CrossRef
  66. Graham AM, Rasmussen JM and Rudolph MD et al (2018) Maternal systemic interleukin-6 during pregnancy is associated with newborn amygdala phenotypes and subsequent behavior at 2 years of age. Biol Psychiatry 83, 109-119.
    Pubmed KoreaMed CrossRef
  67. Northrup TF, Stotts AL and Suchting R et al (2021) Thirdhand smoke associations with the gut microbiomes of infants admitted to a neonatal intensive care unit: an observational study. Environ Res 197, 111180.
    Pubmed KoreaMed CrossRef
  68. McLean C, Jun S and Kozyrskyj A (2019) Impact of maternal smoking on the infant gut microbiota and its association with child overweight: a scoping review. World J Clin Pediatr 15, 341-349.
    Pubmed CrossRef
  69. Palmer C, Bik EM, DiGiulio DB, Relman DA and Brown PO (2007) Development of the human infant intestinal microbiota. PLoS Biol 5, e177.
    Pubmed KoreaMed CrossRef
  70. Beck S, Wojdyla D and Say L et al (2010) The worldwide incidence of preterm birth: a systematic review of maternal mortality and morbidity. Bull World Health Organ 88, 31-38.
    Pubmed KoreaMed CrossRef
  71. Jia Q, Yu X and Chang Y et al (2022) Dynamic changes of the gut microbiota in preterm infants with different gestational age. Front Microbiol 13, 2259.
    Pubmed KoreaMed CrossRef
  72. Moles L, Gomez M and Heilig H et al (2013) Bacterial diversity in meconium of preterm neonates and evolution of their fecal microbiota during the first month of life. PloS One 8, e66986.
    Pubmed KoreaMed CrossRef
  73. Gómez M, Moles L and Espinosa-Martos I et al (2017) Bacteriological and immunological profiling of meconium and fecal samples from preterm infants: a two-year follow-up study. Nutrients 9, 1293.
    Pubmed KoreaMed CrossRef
  74. Forsgren M, Isolauri E, Salminen S and Rautava S (2017) Late preterm birth has direct and indirect effects on infant gut microbiota development during the first six months of life. Acta Paediatr 106, 1103-1109.
    Pubmed KoreaMed CrossRef
  75. Feehily C, Crosby D and Walsh CJ et al (2020) Shotgun sequencing of the vaginal microbiome reveals both a species and functional potential signature of preterm birth. NPJ Biofilms Microbiomes 6, 50.
    Pubmed KoreaMed CrossRef
  76. Stewart CJ, Marrs EC and Nelson A et al (2013) Development of the preterm gut microbiome in twins at risk of necrotising enterocolitis and sepsis. PloS One 8, e73465.
    Pubmed KoreaMed CrossRef
  77. Mai V, Torrazza RM and Ukhanova M et al (2013) Distortions in development of intestinal microbiota associated with late onset sepsis in preterm infants. PloS One 8, e52876.
    Pubmed KoreaMed CrossRef
  78. Bäckhed F, Roswall J and Peng Y et al (2015) Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 17, 690-703.
    Pubmed CrossRef
  79. Arboleya S, Binetti A and Salazar N et al (2012) Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol 79, 763-772.
    Pubmed CrossRef
  80. Liu J, Li Y and Feng Y et al (2019) Patterned progression of gut microbiota associated with necrotizing enterocolitis and late onset sepsis in preterm infants: a prospective study in a Chinese neonatal intensive care unit. PeerJ 7, e7310.
    Pubmed KoreaMed CrossRef
  81. Ho TT, Groer MW and Kane B et al (2018) Dichotomous development of the gut microbiome in preterm infants. Microbiome 6, 1-13.
    Pubmed KoreaMed CrossRef
  82. Korpela K, Blakstad EW and Moltu SJ et al (2018) Intestinal microbiota development and gestational age in preterm neonates. Sci Rep 8, 2453.
    Pubmed KoreaMed CrossRef
  83. Henderickx JG, Zwittink RD, Van Lingen RA, Knol J and Belzer C (2019) The preterm gut microbiota: an inconspicuous challenge in nutritional neonatal care. Front Cell Infect Microbiol 9, 85.
    Pubmed KoreaMed CrossRef
  84. Korpela K (2021) Impact of delivery mode on infant gut microbiota. Ann Nutr Metab 77, 11-19.
    Pubmed CrossRef
  85. Stokholm J, Blaser MJ and Thorsen J et al (2018) Maturation of the gut microbiome and risk of asthma in childhood. Nat commun 9, 141.
    Pubmed KoreaMed CrossRef
  86. Stewart C, Marrs E and Magorrian S et al (2012) The preterm gut microbiota: changes associated with necrotizing enterocolitis and infection. Acta Paediatr 101, 1121-1127.
    Pubmed CrossRef
  87. Beghetti I, Barone M and Turroni S et al (2022) Early-life gut microbiota and neurodevelopment in preterm infants: any role for Bifidobacterium?. Eur J Pediatr 181, 1773-1777.
    Pubmed CrossRef
  88. Hiltunen H, Hanani H and Luoto R et al (2021) Preterm infant meconium microbiota transplant induces growth failure, inflammatory activation, and metabolic disturbances in germ-free mice. Cell Rep Med 2, 100447.
    Pubmed KoreaMed CrossRef
  89. Shaterian N, Abdi F, Ghavidel N and Alidost F (2021) Role of cesarean section in the development of neonatal gut microbiota: a systematic review. Open Med 16, 624-639.
    Pubmed KoreaMed CrossRef
  90. Mitchell CM, Mazzoni C and Hogstrom L et al (2020) Delivery mode affects stability of early infant gut microbiota. Cell Rep Med 1, 100156.
    Pubmed KoreaMed CrossRef
  91. Roswall J, Olsson LM and Kovatcheva-Datchary P et al (2021) Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe 29, 765-776.
    Pubmed CrossRef
  92. Stewart CJ, Embleton ND and Clements E et al (2017) Cesarean or vaginal birth does not impact the longitudinal development of the gut microbiome in a cohort of exclusively preterm infants. Front Microbiol 8, 1008.
    Pubmed KoreaMed CrossRef
  93. Eidelman AI, Schanler RJ and Johnston M et al (2012) Breastfeeding and the use of human milk. Pediatrics 129, e827-e841.
    Pubmed CrossRef
  94. Chong HY, Tan LTH and Law JWF et al (2022) Exploring the potential of human milk and formula milk on infants' gut and health. Nutrients 14, 3554.
    Pubmed KoreaMed CrossRef
  95. Verduci E, Di Profio E and Cerrato L et al (2020) Use of soy-based formulas and cow's milk allergy: lights and shadows. Front Pediatr 8, 591988.
    Pubmed KoreaMed CrossRef
  96. Fabiano V, Indrio F and Verduci E et al (2021) Term infant formulas influencing gut microbiota: an overview. Nutrients 13, 4200.
    Pubmed KoreaMed CrossRef
  97. Huang J, Zhang Z and Wu Y et al (2018) Early feeding of larger volumes of formula milk is associated with greater body weight or overweight in later infancy. Nutr J 17, 12.
    Pubmed KoreaMed CrossRef
  98. Pärnänen KMM, Hultman J and Markkanen M et al (2022) Early-life formula feeding is associated with infant gut microbiota alterations and an increased antibiotic resistance load. Am J Clin Nutr 115, 407-421.
    Pubmed KoreaMed CrossRef
  99. Martin CR, Ling PR and Blackburn GL (2016) Review of infant feeding: key features of breast milk and infant formula. Nutrients 8, 279.
    Pubmed KoreaMed CrossRef
  100. Stewart CJ, Ajami NJ and O'Brien JL et al (2018) Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 562, 583-588.
    Pubmed KoreaMed CrossRef
  101. Lugli GA, Mancabelli L and Milani C et al (2023) Comprehensive insights from composition to functional microbe-based biodiversity of the infant human gut microbiota. NPJ Biofilms Microbiomes 9, 25.
    Pubmed KoreaMed CrossRef
  102. Cong X, Judge M and Xu W et al (2017) Influence of feeding type on gut microbiome development in hospitalized preterm infants. Nurs Res 66, 123-133.
    Pubmed KoreaMed CrossRef
  103. Galazzo G, van Best N and Bervoets L et al (2020) 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 158, 1584-1596.
    Pubmed CrossRef
  104. Korpela K, Helve O and Kolho KL et al (2020) Maternal fecal microbiota transplantation in cesarean-born infants rapidly restores normal gut microbial development: a proof-of-concept study. Cell 183, 324-334.
    Pubmed CrossRef
  105. Masi AC, Embleton ND and Lamb CA et al (2021) Human milk oligosaccharide DSLNT and gut microbiome in preterm infants predicts necrotising enterocolitis. Gut 70, 2273-2282.
    Pubmed KoreaMed CrossRef
  106. Aceti A, Maggio L and Beghetti I et al (2017) Probiotics prevent late-onset sepsis in human milk-fed, very low birth weight preterm infants: systematic review and meta-analysis. Nutrients 9, 904.
    Pubmed KoreaMed CrossRef
  107. Wilson BC, Butler ÉM and Grigg CP et al (2021) Oral administration of maternal vaginal microbes at birth to restore gut microbiome development in infants born by caesarean section: a pilot randomised placebo-controlled trial. EBioMedicine 69, 103443.
    Pubmed KoreaMed CrossRef
  108. Dominguez-Bello MG, De Jesus-Laboy KM and Shen N et al (2016) Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat Med 22, 250-253.
    Pubmed KoreaMed CrossRef
  109. Li X, Li X and Shang Q et al (2017) Fecal microbiota transplantation (FMT) could reverse the severity of experimental necrotizing enterocolitis (NEC) via oxidative stress modulation. Free Radic Biol Med 108, 32-43.
    Pubmed CrossRef
  110. Brunse A, Martin L and Rasmussen TS et al (2019) Effect of fecal microbiota transplantation route of administration on gut colonization and host response in preterm pigs. Isme j 13, 720-733.
    Pubmed KoreaMed CrossRef
  111. Hill C (2020) Balancing the risks and rewards of live biotherapeutics. Nat Rev Gastroenterol Hepatol 17, 133-134.
    Pubmed CrossRef
  112. Bibbò S, Settanni CR and Porcari S et al (2020) Fecal microbiota transplantation: screening and selection to choose the optimal donor. J Clin Med 9, 1757.
    Pubmed KoreaMed CrossRef
  113. Chang CM, Tsai MH and Liao WC et al (2022) Effects of probiotics on gut microbiomes of extremely preterm infants in the neonatal intensive care unit: a prospective cohort study. Nutrients 14, 3239.
    Pubmed KoreaMed CrossRef
  114. Beck LC, Masi AC and Young GR et al (2022) Strain-specific impacts of probiotics are a significant driver of gut microbiome development in very preterm infants. Nat Microbiol 7, 1525-1535.
    Pubmed KoreaMed CrossRef
  115. Stinson LF and Geddes DT (2022) Microbial metabolites: the next frontier in human milk. Trends Microbiol 30, 408-410.
    Pubmed CrossRef
  116. Bering SB (2018) Human milk oligosaccharides to prevent gut dysfunction and necrotizing enterocolitis in preterm neonates. Nutrients 10, 1461.
    Pubmed KoreaMed CrossRef
  117. Alliet P, Puccio G and Janssens E et al (2016) Term infant formula supplemented with human milk oligosaccharides (2'Fucosyllactose and Lacto-neotetraose) shifts stoom microbiota and metabolic signatures closer to that of the breastfed infant. J Pediatr Gastroenterol Nutr 63, S55.
    CrossRef
  118. Kostopoulos I, Elzinga J and Ottman N et al (2020) Akkermansia muciniphila uses human milk oligosaccharides to thrive in the early life conditions in vitro. Sci Rep 10, 14330.
    Pubmed KoreaMed CrossRef
  119. Palleja A, Mikkelsen KH and Forslund SK et al (2018) Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol 3, 1255-1265.
    Pubmed CrossRef
  120. Gasparrini AJ, Wang B and Sun X et al (2019) Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat Microbiol 4, 2285-2297.
    Pubmed KoreaMed CrossRef
  121. Thänert R, Sawhney SS, Schwartz DJ and Dantas G (2022) The resistance within: antibiotic disruption of the gut microbiome and resistome dynamics in infancy. Cell Host Microbe 30, 675-683.
    Pubmed KoreaMed CrossRef
  122. Dibo M, Ventimiglia MS, Valeff N, Serradell M and Jensen F (2022) An overview of the role of probiotics in pregnancy-associated pathologies with a special focus on preterm birth. J Reprod Immunol 150, 103493.
    Pubmed CrossRef
  123. Hui Y, Smith B and Mortensen MS et al (2021) The effect of early probiotic exposure on the preterm infant gut microbiome development. Gut Microbes 13, 1951113.
    Pubmed KoreaMed CrossRef
  124. Asbury MR, Shama S and Sa JY et al (2022) Human milk nutrient fortifiers alter the developing gastrointestinal microbiota of very-low-birth-weight infants. Cell Host Microbe 30, 1328-1339.
    Pubmed CrossRef
  125. Park S, Belfoul AM and Rastelli M et al (2023) Maternal low-calorie sweeteners consumption rewires hypothalamic melanocortin circuits via a gut microbial co-metabolite pathway. JCI Insight 8, e156397.
    Pubmed KoreaMed CrossRef
  126. Roager HM, Stanton C and Hall LJ (2023) Microbial metabolites as modulators of the infant gut microbiome and host-microbial interactions in early life. Gut Microbes 15, 2192151.
    Pubmed KoreaMed CrossRef
  127. Paredes A, Justo-Méndez R and Jiménez-Blasco D et al (2023) γ-Linolenic acid in maternal milk drives cardiac metabolic maturation. Nature 618, 365-373.
    Pubmed CrossRef
  128. Liang L, Rasmussen MH and Piening B et al (2020) Metabolic dynamics and prediction of gestational age and time to delivery in pregnant women. Cell 181, 1680-1692.
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Author ORCID Information

Funding Information

Collections

Services
Social Network Service

e-submission

Archives